Next Article in Journal
Revisiting Resveratrol as an Osteoprotective Agent: Molecular Evidence from In Vivo and In Vitro Studies
Next Article in Special Issue
Fibroblast Growth Factor 23: Potential Marker of Invisible Heart Damage in Diabetic Population
Previous Article in Journal
Association between Organochlorine Pesticides and Vitamin D in Female Subjects
Previous Article in Special Issue
Microbiota Metabolism Failure as a Risk Factor for Postoperative Complications after Aortic Prosthetics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion

1
Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
2
Internal Medicine Department, Zaporozhye State Medical University, 69035 Zaporozhye, Ukraine
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(5), 1452; https://doi.org/10.3390/biomedicines11051452
Submission received: 31 March 2023 / Revised: 25 April 2023 / Accepted: 25 April 2023 / Published: 16 May 2023

Abstract

:
Atrial fibrillation (AF) is associated with atrial remodeling, cardiac dysfunction, and poor clinical outcomes. External direct current electrical cardioversion is a well-developed urgent treatment strategy for patients presenting with recent-onset AF. However, there is a lack of accurate predictive serum biomarkers to identify the risks of AF relapse after electrical cardioversion. We reviewed the currently available data and interpreted the findings of several studies revealing biomarkers for crucial elements in the pathogenesis of AF and affecting cardiac remodeling, fibrosis, inflammation, endothelial dysfunction, oxidative stress, adipose tissue dysfunction, myopathy, and mitochondrial dysfunction. Although there is ample strong evidence that elevated levels of numerous biomarkers (such as natriuretic peptides, C-reactive protein, galectin-3, soluble suppressor tumorigenicity-2, fibroblast growth factor-23, turn-over collagen biomarkers, growth differential factor-15) are associated with AF occurrence, the data obtained in clinical studies seem to be controversial in terms of their predictive ability for post-cardioversion outcomes. Novel circulating biomarkers are needed to elucidate the modality of this approach compared with conventional predictive tools. Conclusions: Biomarker-based strategies for predicting events after AF treatment require extensive investigation in the future, especially in the presence of different gender and variable comorbidity profiles. Perhaps, a multiple biomarker approach exerts more utilization for patients with different forms of AF than single biomarker use.

1. Introduction

Atrial fibrillation (AF) is the most common form of sustained cardiac arrhythmia in the world [1]. The prevalence of AF advances with increasing age. After the age of 80, atrial fibrillation affects 10–17% of the population [2]. The morbidity is increased and mortality rises up to 3.5-fold in men and women [3]. Along with it, AF frequently occurs in patients at higher risk of cardiovascular diseases (CVD) as well as among individuals with known CVD [4]. Unfortunately, AF and CVD exacerbate each other and mutually intervene in prognosis. Indeed, patients with any form of AF demonstrated poorer clinical outcomes if there is concomitant heart failure (HF), coronary artery disease (CAD), type 2 diabetes mellitus (T2DM), obesity, obstructive sleep apnea, chronic kidney disease (CKD), or peripheral artery disease [5,6,7]. Further, the prognosis of patients with AF is poorer than the prognosis of patients with various CVD and comorbid conditions (i.e., HF, CKD) without AF [8]. Multi-morbidity among patients with AF seems to play a pivotal role in natural evolution of primary and secondary AF through direct and indirect impact on the structural and/or electrophysiological abnormalities that occur in AF [9,10]. AF influences electrical remodeling, i.e., shortening of refractoriness due to the high atrial rate itself, resulting in adverse cardiac remodeling [11]. Yet, the persistence of AF itself modulates the risk of cerebrovascular and cardiovascular events [12,13].
The management of AF includes either rhythm restoration or rate control along with comorbidity management, prevention of stroke, and systemic thromboembolism [14]. Synchronized electrical cardioversion can terminate AF. Combined with sedation, it is a safe procedure and highly effective, restoring sinus rhythm in more than 90% [15,16,17]. It is important to detect AF recurrence after successful electrical cardioversion. In this case, early cardioversion could prolong the subsequent duration of sinus rhythm and slow disease progression compared to delayed sinus rhythm restoration [18].
Although the current clinical protocol of initial AF management seems to be very useful in practice [1], it poses challenges in predicting incidental AF and early detection of AF-related complications [19,20]. There are many factors associated with AF recurrence, such as duration of AF, higher age, sex, HF, LA volume index, chronic obstructive pulmonary disease, hypertension, obstructive sleep apnea, hyperthyroidism, smoking, and obesity [21,22]. However, the role of biomarkers reflecting the different stages of AF pathogenesis has not been completely understood. The purpose of the study is to summarize the current evidence on the value of various biomarkers in predicting the likelihood of AF recurrence after electrical cardioversion.

2. Promoting Factors and Electrophysiological/Anatomical Substrates of AF

Vulnerable substrates for the occurrence, support, and recurrence of AF are electrophysiological and adverse cardiac remodeling, along with structural remodeling, mechanical dysfunction, and trigger activity, which are mediated by genetic ion channel alterations, concomitant cardiovascular (CV) diseases (acute and chronic coronary syndromes, multifocal atherosclerosis, primary and secondary cardiomyopathy, etc.), CV risk factors (hypertension, smoking, obesity, diabetes mellitus, resistance to insulin, and dyslipidemia), and comorbidities (chronic obstructive pulmonary disease, bronchial asthma, chronic kidney disease) (Figure 1). In addition, concomitant hemodynamic factors as a result of numerous diseases (heart failure, atrial cardiomyopathy, pulmonary hypertension, inherited and acquired heart diseases, myocarditis) and conditions (chemotherapy, cardiac toxicity) play a crucial role in secondary structural remodeling of the heart [23,24,25]. These factors contribute to AF occurrence by maintaining afterdepolarization-induced triggered ectopic activity, focal enhanced automaticity, altered function of ion channels, micro-reentrant circus rotor, less dynamic head–tail interactions during re-entry in cardiac tissue, altered ion accumulation on the dynamics of re-entry and electrical heterogeneity [26]. Indeed, head–tail interactions have previously been known to have a causative impact on the dynamics of the reentrant action potential, which plays a pivotal role in inducing AF [26]. To note, intracellular ions, mainly Ca2+ and Na+, accumulated during reentrant arrhythmia through the rapid repetitive cellular excitation may lead to spontaneous termination of re-entry or break-up of the re-entry loop into multiple pathways resulting in AF. Along with it, the initiation and persistence of AF are controlled by both parasympathetic and sympathetic stimulation, as well as hormonal influences, which also seem to play a role in AF recurrence [27]. However, the continuous interaction between electrophysiological, structural, and anatomical remodeling leads to intercellular uncoupling and a pro-fibrotic response, which is crucial for trigger activity, the presence of AF, and the transformation of cardiac dysfunction into HF [28].

2.1. Electrophysiological Remodeling

Electrophysiological remodeling affects variable changes in specific ionic currents, such as a reduction in transient outward potassium current, L-type calcium current, and ultra-rapid delayed rectifier current, as well as shortening of the effective refractory period and prolongation of the action potential, which are also associated with the increase in the stimulation rate [29,30]. The overload of intracellular calcium in cardiac myocytes and its spontaneous release from the sarcoplasmic reticulum seems to be a major factor in the occurrence of delayed afterdepolarizations and triggered ectopic activity in the myocardium [30]. Although sympathetic activation and direct stimulation by angiotensin-II are classic mechanisms of enhancing propensity for AF, there are numerous other mechanisms that intervene in altered afterdepolarizations. They mainly include a reduced inward rectifier, as well as increased activity of the Na/Ca exchanger and residual beta-adrenergic responsiveness [31,32]. Yet, alterations in the regulation and accumulation of intracellular calcium can be a result of properly persistent AF and alternative arrhythmogenic mechanisms (intramural decremented conduction, transmural heterogeneity of repolarization, prolongation of QT-interval, and block of the premature impulse), which are activated due to progress of pre-exciding CV diseases including HF and coronary artery syndromes [33]. To note, asynchronous down-regulation of voltage-dependent potassium currents and L-type calcium currents between layers of myocardium through the calcium/calmodulin-dependent kinase II signal pathway activated by hemodynamic factors (fluid overload, hypertension), ischemia/hypoxia, hormonal dysfunction (hyperthyroidism), perivascular edema due to microvascular inflammation, impaired mitochondrial metabolism and oxidative stress due to metabolic diseases/conditions (diabetes mellitus, obesity, myopathy, insulin resistance) and cardiac hypertrophy may support electrophysiological remodeling [34,35,36,37,38,39]. Although the role of hemodynamic factors and ischemia in shaping AF risk is well established [34,35], the impact of metabolic influences on electrophysiological remodeling is not always obvious. For instance, among patients with thyroid dysfunction, free thyroxine levels but not thyroid-stimulating hormone concentrations are associated with an increased risk of incident AF regardless of preexisting CV disease [37]. On the other hand, a hypothyroid state may directly induce myocardial fibrosis via stimulating autophagy and inhibiting TGF-β1/Smad2 signal transduction pathway [38]. Obesity and T2DM link glycemic fluctuations to electrophysiological remodeling that leads to the onset and maintenance of AF through mitochondrial dysfunction, oxidative stress, and inflammation [39]. Chan YH et al. (2019) [40] reported that insulin resistance (IR) was associated with significantly increased sarcoplasmic reticulum calcium content and diastolic calcium sparks in the atrial myocardium. Moreover, IR increased collagen accumulation and superoxide production in the atrial myocardium through increased synthesis of transforming growth factor beta 1 (TGF-β1) and abnormal upregulation of calcium-homeostasis-related proteins, such as oxidized CaMKIIδ, phosphorylated-phospholamban, phosphorylated-RyR-2, and sodium–calcium exchanger [40].
Yet, subcellular mechanisms underlying electrophysiological remodeling seem to relate to the alteration of connexin 43 expression, which is a principal ventricular gap junction protein [40,41]. However, significant changes in connexin 43 phosphorylation were found to be more closely associated with timing AF persistence and the presence of HF [42]. In particular, these changes can even explain an association of such powerful components of electrophysiological features as increased transmural dispersion in refractoriness and conduction with increased inducibility of AF and low efficacy of electrical cardioversion [43,44]. Moreover, this may be a novel paradigm of electrophysiological remodeling based on the timing of conduction abnormalities in connection to dynamic changes in connexin 43 isoforms, cardiac dysfunction, and comorbidities [43,44]. Indeed, there is strong evidence of the fact that apelin-13-an aliphatic multifunctional peptide, mainly originated from the myocardium, skeletal muscles, and liver-increased connexin 43 through autophagy inhibition and inducing AKT and mTOR phosphorylation and thereby decreases susceptibility to cardiac arrhythmias including AF and cardiomyocyte death [45,46]. Yang M et al. (2022) [47] recently reported that the apelin/AMPK/mTOR signaling pathway, which regulates angiotensin II-mediated autophagy and apoptosis of cardiac myocytes, is under close control of miRNA-122-5p. The overexpression of miRNA-122-5p leads to exacerbation of cardiac and vascular hypertrophy, cardiac fibrosis, and dysfunction. Thus, the overexpression of miRNA-122-5p may be an underlying mechanism of binding myocardial fibroblasts and activation of AF. On the other hand, angiotensin-II acts as a promoter of expression of pro-apoptotic molecules, such as P62 and Bax, and as a mediator of mTOR phosphorylation, which downregulates LC3II, beclin-1, and contributes to the imbalance of autophagy and apoptosis in the myocardium. These changes were associated with increased myocardial accumulation of collagen I and collagen III, overexpression of TGF-beta-1 and connective tissue growth factor (CTGF), as well as downregulation of myocardial expression of apelin, angiotensin-converting enzyme-2 (ACE2), and growth differential factor-15 (GDF-15) [47,48,49]. These facts confirm a close interplay between the Apelin-APJ axis and ACE2-GDF-15-porimin signaling in angiotensin-II-mediated myocardial hypertrophy and fibrosis, which are crucial substrates for AF occurrence and prolongation. Therefore, they modulate the relationship between electrophysiological and anatomical cardiac remodeling.

2.2. Adverse Cardiac Remodeling

Adverse cardiac remodeling in AF patients includes AF-related atrial remodeling and cardiac remodeling due to concomitant CV diseases [50]. Both variants may be associated with sinus node dysfunction, variability in conduction gaps due to parasympathetic/sympathetic stimulation and epigenetic regulation of intercellular communication, cardiac cell-to-cell heterogeneity, and extracellular matrix alteration [50,51,52,53]. Therefore, the overlap between both variants of remodeling is mediated by concomitant hemodynamic changes such as valvular regurgitation [54]. AF-related alteration of atrial structure starts with the differentiation of cardiac fibroblasts into myofibroblasts, which is regulated by numerous triggers, including angiotensin-II, noradrenaline, thyroid hormones, inflammatory cytokines, chemokines, matrix metalloproteinases, galectine-3, soluble suppression of tumorigenesis-2 (sST2), TGF-beta-1 and microRNAs (Figure 2).
These triggers contribute to the altered expression of several ion channel proteins, such as transient receptor potential channel-3 (TRPM3) and member 7 TRPC3, which regulate intracellular calcium flow, and mediate dysfunction of the ion channels on the surfaces of target cells [54]. Angiotensin-II, aldosterone, endothelin, and catecholamines, as well as several inflammatory cytokines (TNF-alpha, interleukin-2) acting as signaling molecules contribute to fibroblast proliferation via Ca2+ entry via transient receptor potential channels (voltage-gated sodium [Nav1.5] and potassium channels [Kv1.5]) [55]. In addition, myofibril protein breakdown is stimulated by overexpressed calpain, which is activated by intracellular Ca2+ loading [56]. As a result, activated myofibroblasts not only produce several types of collagens shaping collagen deposition and cardiac fibrosis but also directly interact with cardiomyocytes promoting AF [57]. Moreover, myofibroblasts and fibrotic areas interfere with atrial tissue conduction and lead to intercellular uncoupling. Indeed, interactions between myofibroblast and cardiomyocyte alter conduction and elicit focal activity in the atria [58]. Finally, cell uncoupling, along with cardiac myocyte disarmament and extensive fibrosis, lead to gap junctions and non-uniformity of anisotropy modulating AF. In addition, pre-exceeding CV diseases, such as myocardial infarction, cardiomyopathies, myocarditis, and cardiac hypertrophy, through the strength of local mechanical forces, loss of cardiac myocytes and extensive fibrosis intervene in anisotropy modality of cardiac electrical conductivity and shaping arrhythmogenic substrate [59].
Although collagen accumulation in the myocardium is regulated by autocrine/paracrine and neurohumoral mechanisms, the atria are more prone to extracellular matrix remodeling and collagen deposition than the ventricles. Possibly, it depends on the distinguished presence of matrix metalloproteinases, their inhibitors, and pro-inflammatory molecules involved in the subsequent regulation of collagen synthesis and degradation. The accumulation of collagen, other matrix proteins (elastin, fibronectin 1, fibrillin 1), and proteoglycans in abundance lead to severe heterogeneous areas in the atria with variable alteration of electrophysiological properties [59]. This eventually leads to changes in myocardial cell architecture, such as elongation and disturbed alignment of demarcated fibers. This subsequently causes anisotropic changes in the entire myocardium, mediating a discrepancy between transverse and longitudinal electrical conduction leading to AF.

3. Electrical Cardioversion of AF: Safety and Outcomes

It seems that standard external direct current electrical cardioversion is a well-developed urgent treatment strategy for patients presenting recent-onset AF [1,60]. Numerous retrospective one-center studies and multicenter trials yielded 86–88% efficacy of the approach in restoring sinus rhythm along with 6–10% relapse of AF in a short-term perspective (7–28 days) [61,62,63]. Overall, electrical cardioversion in AF patients who required emergency department transportation was associated with infrequent hospital admission and few mild-to-moderate complications [61]. However, the duration of AF in the majority of studies was less than 48 h in 99% of the patients. Burton JH et al. (2004) [61] observed in a retrospective multicenter study that electrical cardioversion had an 86% success rate, and only 10% of the patients returned to the emergency department within 7 days. Fried AM et al. (2021) [16] reported that the efficacy of this procedure, defined as restoration of sinus rhythm, reached 88% in routine clinical practice, whereas major complications (post-cardioversion stroke, thromboembolic events, jaw thrust maneuver for hypoxia, and overnight observation for hypotension) and predefined minor adverse events (frequently related to general anesthesia, skin burns) were detected in 0.3% and 14%, respectively. In addition, electrical cardioversion was about 2.5 times more effective than conventional pharmacological treatment in restoring sinus rhythm [62,63]. Although there are numerous potential complications of electrical cardioversion (i.e., ventricular fibrillation, thromboembolism due to inadequate anticoagulant therapy, nonsustained ventricular tachycardia, various forms of atrial arrhythmias, bradycardia, transient left bundle branch block, myocardial necrosis, asymptomatic myocardial dysfunction, acute HF, transient hypotension, pulmonary edema, and stroke), they occur less frequently than recurrent AF. Further, 6.4% of patients revisited the emergency department within 30 days, and 4.8% returned with AF or atrial flutter. It is noteworthy that the return visit rate for patients with relapsed AF varies between 3% and 17% [64].
Overall, 30-day all-cause mortality among AF patients undergoing direct-current electrical cardioversion was 0.8% [65]. Data received from the FIRE (Atrial Fibrillation/flutter Italian Registry) registry showed that predictors of unsuccessful electrical cardioversion were onset of AF > 48 h, concomitant HF, increasing age, syncope, transient ischemic attack (TIA)/stroke as well as previous admission to a non-cardiology department [66]. The investigators also found several predictors of in-hospital mortality in this patient population, including age, HF, diabetes mellitus, previous admission to a non-cardiology department, and TIA/stroke [66]. Thus, patients at low risk for thromboembolic complications, including stroke and heart failure, seem to benefit more from electric cardioversion than other individuals with recent-onset AF [67].
Another reason for physicians to use this approach may be cost savings and a short period of emergency department admission [67,68]. Houghton AR et al. (2000) [69] and Boriani G. et al. (2007) [70] did not identify concise hemodynamic predictors of successful external electrical cardioversion or relapses after electrical cardioversion among patients with persistent AF or atrial flutter. However, only two predictors (duration of arrhythmia ≥1 year and previous cardioversion) were found to be powerful for this matter [69,70], whereas, in previous investigations, relapse of AF was associated with reduced left ventricular ejection fraction [71]. Along with it, standard external biphasic direct current electrical cardioversion has better efficacy than monophasic electrical cardioversion (360-J) for restoration of sinus rhythm in AF patients, although dual external monophasic 360-J cardioversion may increase the success rate as a rescue technique after failing standard external direct current cardioversion [72,73]. In this concept, the prediction of plausible cardiovascular events, including relapsed AF, with a biomarker strategy seems promising in patients with recent-onset AF.

4. Predictors for AF Recurrence Following Electrical Cardioversion

Biomarkers reflecting the complex pathophysiological mechanisms underlying AF seem to be an effective tool to predict rhythm status after cardioversion as well as other AF-related complications, which can intervene in mortality, hospital admission, cardiovascular (CV), and non-CV outcomes (Table A1).

4.1. Natriuretic Peptides

Natriuretic peptides (brain natriuretic peptide [BNP], N-terminal pro-B-type natriuretic peptide (NT-proBNP), mid-regional pro-A type natriuretic peptide (MR-proANP)) serve as circulating cardiac biomarkers of biomechanical stress, adverse cardiac remodeling and fluid overload with established diagnostic and predictive values for acute and chronic HF involving any phenotypes [74,75]. Along with it, elevated levels of NPs were strongly associated with all-cause and CV mortality and urgent hospitalization among patients with AF, T2DM, CKD, hypertension, and cardiac hypertrophy [76,77]. Moreover, NT-proBNP and BNP were found to be predictors for AF [78,79,80]. However, it has been suggested that restoration of sinus rhythm through effective electric cardioversion may associate with a reduction in NP concentrations and thereby predict the recurrence of new episodes of arrhythmia. Xu X et al. (2017) [81] observed in a meta-analysis that low levels of BNP and NT-proBNP were associated with the maintenance of sinus rhythm and that the baseline concentrations of both biomarkers may be a predictor of AF recurrence after successful electrical cardioversion. Ari H. et al. (2008) [82] reported that a significant decrease in BNP levels 30 min after electric cardioversion corresponded to six-month maintenance of sinus rhythm in follow-up.
In the GAPP-AF (The gene expression patterns for the prediction of atrial fibrillation) study, Meyre PB (2022) [83] investigated 21 conventional and new circulating biomarkers reflecting inflammation, myocardial injury, cardiac biomechanical stress, and renal dysfunction before and 30 days after electrical cardioversion and evaluated plausible associations of changes in circulating biomarker levels with rhythm status at 30-day follow-up. The patients included in the study had no acute HF, severe valvular disease, or life-limiting active or chronic serious concomitant diseases. The authors found that low levels of NT-proBNP were independently associated with sinus rhythm restoration after electric cardioversion. On the other hand, initial levels of BNP and NT-proBNP in patients with persistent AF without established CVD did not predict long-term sinus rhythm maintenance, although conversion to sinus rhythm related to a significant decrease in circulating BNP but not NT-proBNP level [84]. In contrast, NT-proBNP levels were found to be a predictor of AF recurrence 30 days after successful electric cardioversion among patients with persistent AF and CV risk factors, including hypertension and dyslipidemia [85]. In another study, pre-procedural NT-proBNP levels, but not post-procedural levels of the peptide, independently predicted the relapse of AF after successful electrical cardioversion [86]. These controversial issues perhaps may relate to the presence of concomitant HF. Indeed, in the CAPRAF (Candesartan in the Prevention of Relapsing Atrial Fibrillation) trial, plasma NT-proBNP concentrations measured before electrical cardioversion did not predict cardioversion success nor the relapse of AF in patients without HF [87]. Mabuchi N et al. (2000) [88] noticed that low atrial natriuretic peptide (ANP) and high BNP levels before electric cardioversion were independent predictors of recurrent AF in mild chronic HF patients. Moreover, the authors established that ANP to BNP ratio <0.44 was a significant risk factor for AF recurrence [88]. The BNP level of 700 fmol/mL or higher on day 7 after cardioversion was most predictive for AF recurrence (sensitivity, 78%; specificity, 71%), whereas ANP did not predict the relapse of AF [89]. Buccelletti F. et al. (2011) [90] measured the levels of NT-proBNP in 200 patients admitted to the emergency department due to new-onset AF (<2 weeks) regardless of HF presence. The authors found that NT-proBNP levels of either ≤450 pg/mL or >1800 pg/mL seem to show positive and negative predictive values for cardioversion in rate-control and rhythm-control strategies, respectively. In the range of 450 to 1800 pg/mL, NT-proBNP did not exhibit serious clinical utility [90]. However, it remained unclear whether continuous monitoring of the dynamic changes of NPs after sinus rhythm restoration predicts recurrent AF [91]. Overall, the restoration of sinus rhythm after electric cardioversion in AF patients is associated with a decrease in circulating levels of NPs and low levels of NT-proBNP predicts a sustainable maintains of sinus rhythm in follow-up.

4.2. Biomarkers of Fibrosis

Cardiac fibrosis was found to be closely associated with AF. Circulating biomarkers of fibrosis have already been proposed as a promising tool in its evaluation, but which biomarkers are most appropriate for AF remains unclear [92]. There are a large number of circulating biomarkers, which characterize the accumulation of extracellular matrix components and fibrosis, such as soluble suppressor tumorigenicity-2 (sST2), galectin-3 (Gal-3), procollagen type III N terminal peptide (PIIINP), type I collagen carboxyl telopeptide (ICTP), and fibroblast growth factor 23 (FGF-23) [93].

4.2.1. Galectin-3

Gal-3 is a multifunctional galactose-binding protein that belongs to the transforming growth factor beta superfamily and a biomarker of fibrosis, involved in atrial remodeling, cardiac fibrosis, and AF [94]. Previous studies revealed that patients with AF had higher Gal-3 values than non-AF patients, regardless of their comorbidity profile [95,96]. Moreover, elevated Gal-3 levels were independently associated with paroxysmal non-valvular AF [97].
There is ample evidence of a close relation between elevated Gal-3 levels, atrial remodeling (i.e., parameters of left atrial dimension, volume, compliance, and contractility) and AF recurrence following successful electrical cardioversion [98,99,100]. Gürses KM et al. (2019) [98] reported that pre-cardioversion Gal-3 levels in persistent AF corresponded to a higher left atrial volume index and were associated with early AF recurrence following successful sinus rhythm restoration. In contrast, Cichoń M et al. (2021) [101] did not find any link between circulating Gal-3 levels and the risk of recurrent AF in obese and non-obese patients with persistent AF. The same results were obtained in another study involving 75 non-HF patients with paroxysmal or persistent AF referred for electrical cardioversion [102]. Although the authors of the study established a correlation between the Gal-3 levels and oxidative stress and inflammation in AF patients, only circulating myeloperoxidase, but not Gal-3, was associated with the maintenance of sinus rhythm in a multivariate model, possibly due to the small number of patients and relatively early stage of AF [102]. Whether these changes may be explained in connection with single nuclear polymorphisms of the Gal-3 gene has not been fully elucidated [103]. Thus, the predictive ability of Gal-3 for sinus rhythm restoration following successful electrical cardioversion requires thorough investigations in face-to-face comparison with other biomarkers before implementation in clinical practice.

4.2.2. sST2

Soluble suppression of Tumorigenicity 2 protein (sST2) is part of the interleukin 1 receptor/Toll-like superfamily, which is related to cardiac inflammation, fibrosis, and also remodeling. Current clinical guidelines for HF consider sST2 as an alternative biomarker of all-cause and CV mortality as well as HF-related complications, including hospital admission, especially in HF with preserved ejection fraction (HFpEF) [74,75]. Although sST2 is involved in cardiac fibrosis, local and systemic inflammation, and atrial and ventricular remodeling, its role in predicting clinical outcomes of electrical cardioversion of AF remains uncertain [104,105]. In patients with HF and acute myocardial infarction, elevated sST2 levels were a powerful risk factor for new-onset AF [106,107]. Moreover, in AF patients without concomitant cardiovascular disease, sST2 concentrations were positively associated with LV myocardial strain and T1 mapping indices [108]. Previous studies have demonstrated significant predictability of AF recurrence after cryoballoon and radiofrequency ablation using sST2 [109,110,111].
It appears that limited evidence exists regarding a discriminatory effect of sST2 measured before and after electrical cardioversion on AF recurrence. Wałek P. et al. (2020) [112] found that sST2, but not Gal-3, predicted sinus rhythm maintenance after successful electrical cardioversion of AF in patients without HF. Perhaps, sST2 may be considered as part of a multimarker panel for the prediction of AF recurrence along with NPs and Gal-3. Overall, sST2 seems to be a promising predictive biomarker for AF recurrence after electrical cardioversion, cryoballoon, and radiofrequency ablation.

4.2.3. Other Biomarkers of Fibrosis

Begg GA (2017) [113] investigated an association of biomarkers related to fibrosis and collagen metabolism with procedural risk and AF recurrence rates among 79 patients undergoing external direct current cardioversion in comparison with 40 age-and-disease-matched volunteers. The authors found that Gal-3, PIIINP, and ICTP were not predictive for AF recurrence after electrical cardioversion, whereas FGF-23 had a weak predictive ability for relapsing AF [113]. In contrast, Kawamura M. et al. (2012) [114] found no discriminatory levels of interleukin-6, high-sensitivity C-reactive protein, BNP, renin, and aldosterone for the 24-month recurrence rate of AF, whereas baseline serum levels of PIIINP > 0.72 U/mL predicted AF relapse. Thus, there is a serious discrepancy between biomarker levels corresponding to the presence of atrial fibrosis confirmed by cardiac magnetic resonance imaging and their discriminatory properties for recurrent AF [115,116,117].
Furthermore, elevated serum levels of FGF-23 strongly correlated with the total number of major cardiovascular events and left atrial dimension in paroxysmal AF patients as well as with new-onset AF in sinus rhythm patients presenting CV risk factors, but not with the maintenance of sinus rhythm during follow-up [117,118,119]. Meta-analysis of 15 clinical studies, enrolling 36,017 participants, revealed that elevated serum FGF-23 levels, but not GDF15 levels, were associated with the risk of AF [120]. A meta-analysis of 15 clinical trials involving 36,017 participants found that elevated serum FGF-23 levels, but not GDF15 levels, were associated with AF risk [120]. However, it remains unclear whether these results also apply to patients undergoing electrical cardioversion.

4.3. Biomarkers of Inflammation

4.3.1. GDF15

GDF15 is a member of the TGF-beta superfamily whose expression is increased in response to biomechanical myocardial stress, inflammation, or ischemia/hypoxia [121]. GDF15 is involved in the regulation of energy homeostasis, thermogenesis, and eating behavior [122]. Yet, GDF15 also exerts anti-inflammatory and anti-proliferative properties, although the underlying molecular mechanisms are still unclear [123]. Elevated GDF15 levels were found in patients with any phenotypes of chronic HF, stroke, AF, and T2DM [124,125,126,127,128]. In the general population, GDF-15 did not show a positive association with the prevalence of AF and the risk of AF occurrence [129]. The suitability of GDF15 for predicting bleeding and/or atrial thrombosis during anticoagulant therapy remains questionable [130]. Clinical evidence for the discriminative value of GDF15 for AF relapse or sinus rhythm maintenance is extremely limited. There is one small study that prospectively included 82 patients with persistent AF [101]. Although log10 serum GDF-15 levels correlated positively with the CHA2DS2-VASc score, there was no close association between GDF-15 levels and sinus rhythm maintenance in patients after successful electric cardioversion [101]. Thus, a discriminative potency of GDF15 for the prediction of clinical efficacy of electrical cardioversion among patients with nonvalvular/valvular AF is not completely understood and requires scrutiny in large clinical studies.

4.3.2. hs-CRP

High-sensitivity C-reactive protein (hs-CRP) is a classic biomarker of inflammation and is a component of the inflammatory profile observed in AF patients. Elevated hs-CRP levels were found in patients with all forms of nonvalvular/valvular AF, regardless of etiology and concomitant comorbidities [131,132]. hs-CRP predicted new-onset AF both in the general population as well as in patients with established cardiovascular or metabolic diseases, such as HF, acute myocardial infarction, T2DM, and metabolic syndrome [133,134,135]. Among patients with AF complicated by systemic thromboembolism, the levels of hs-CRP correlated positively with the CHA2DS2-VASc score [136].
Loricchio ML et al. (2007) [137] investigated plausible predictors for a 1-year risk of AF recurrence after electrical cardioversion. In a Cox regression analysis, the authors found that age, gender, hypertension, T2DM, LVEF, left atrial diameter, use of various antiarrhythmic and antihypertensive (including angiotensin-converting enzyme inhibitors or angiotensin II antagonists) drugs, and statins were not associated with relapsing AF. On the contrary, a low quartile of hs-CRP levels was found to be a strong predictor for this outcome [137]. Lombardi F. et al. (2008) [138] did not find any changes in hs-CRP levels after cardioversion in patients with persistent AF and preserved LVEF, regardless of the post-procedural underlying rhythm. However, NT-proBNP levels decreased significantly in patients who maintained sinus rhythm but not in those who had AF. Yet, baseline hs-CRP levels, but not echocardiographic features of atrial dysfunction and initial NT-proBNP levels, predicted recurrences of AF after cardioversion in patients without pre-existing left ventricular dysfunction [138]. Barassi A et al. (2012) [139] and Korantzopoulos P et al. (2008) [140] confirmed that in patients with persistent AF and preserved LVEF, elevated hs-CRP levels independently predicted subacute AF recurrence rate, whereas NT-proBNP concentrations were not associated with arrhythmic outcome but corresponded to the alterations of cardiac hemodynamics secondary to the presence of AF.
Overall, there is ample strong evidence that elevated preprocedural hs-CRP levels may provide independent predictive information for both successful electrical cardioversion of AF and maintenance of sinus rhythm after conversion [141,142]. The meta-analysis by Liu et al. [143], which included six prospective observational studies (n = 366 patients), showed that peripheral blood CRP levels were higher in patients with failed electric cardioversion than in those with successful restoration of sinus rhythm. In another meta-analysis by Yo CH et al. (2014) [144], a cut-off value of 1.9 mg/L hs-CRP predicted long-term AF recurrence (77% sensitivity, 65% specificity), and more than 3 mg/L predicted short-term AF relapse (73% sensitivity, 71% specificity). Thus, the measurement of CRP levels before the procedure may provide additional prognostic information about the success of sinus rhythm maintenance.
In addition, there are data illustrating that hs-CRP levels measured shortly after electrical cardioversion may be a powerful biomarker for assessing the risk of relapsing AF in the long-term. In particular, Celebi OO et al. (2011) [145] reported that hs-CRP levels measured before and 2 days after electrical cardioversion predicted the 1-year risk of AF relapse. Whether postprocedural hs-CRP provides more information to predict the event than preprocedural hs-CRP is still unclear. However, elevated levels of hs-CRP predicted new-onset AF in the general population and among patients with known cardiovascular diseases, while their role as a marker of sustainable sinus rhythm control places under question.

4.4. Myokines and Adipocytokines

Several interdependent canonic signaling pathways, such as the renin-angiotensin-aldosterone system; TGF-beta pathway, inflammatory chemokines, and cytokines lead to cardiac fibrosis through modulation of oxidative stress and inflammation. However, the direct mechanical stretch may act as a modulator of extracellular matrix remodeling by attenuating the expression of matrix metalloproteinases and their inhibitors. Recently, another signaling pathway has been identified that induces atrial fibrosis via the secretion of adipokines from epicardial, perivascular, and adipose tissue white adipocytes. In addition, recent studies have shown that myokines derived from cardiac and skeletal muscle myocytes may act as adaptive regulators of extracellular matrix remodeling and can attenuate fibrosis [146,147]. Depending on their origin, adipokines and myokines may modulate myofibroblast capabilities, regulate myocyte energy homeostasis and protect against inflammation and fibrosis [148,149]. However, some pro-fibrotic adipokines and myokines can switch a generation of reactive oxygen species to pro-inflammatory and pro-fibrotic stimuli, stimulate myofibroblast differentiation through JAK/STAT3 and JNK/c-Jun signaling, interfere with myocyte electrophysiology, and promote fibrosis in the myocardium [150,151,152]. Numerous previous studies have shown that resistin, apelin, and adiponectin are adipokines associated with several known risk factors for AF and risk of AF [153,154,155,156]. A recent meta-analysis of 34 studies (total number of patients = 31,479) showed that some adipokines, mainly adiponectin, apelin, and resistin, were associated with the risk of AF in the pooled univariate data, whereas the associations were not apparent after multivariate adjustment [157]. However, there is limited evidence of the relation between adipokine and myokine signatures and the risk of AF-related outcomes after electric cardioversion.

4.4.1. Apelin

Apelin is a multifunctional regulatory peptide with potential cytoprotective properties. It is a ligand of the angiotensin II protein J receptor (APJ) receptor and belongs to the G protein-coupled receptor family [158]. Apelin mRNA is widely expressed in tissues such as the cardiovascular, central nervous, adipose, skeletal muscles, and gastrointestinal systems. The Apelin/APJ axis mediates signal transduction for regulating energy homeostasis, including glucose and lipid metabolism, mitochondrial function, angiogenesis, cellular proliferation, and differentiation [159]. Furthermore, apelin inhibits apoptosis, decreases myocardial infarction size, and prevents myocardial ischemia/reperfusion injury via the PI3K/Akt and ERK1/2 caspase signaling. It is also engaged in the autophagy pathway, attenuation of inflammatory reactions, and prevention of atherosclerotic plaque formation [160]. Several controversial issues remain regarding whether the apelin/APJ system is essential for regulating atrial and ventricular remodeling by alleviating myocardial hypertrophy induced by angiotensin II, oxidative stress, and TGF-beta1 [161,162,163]. Nevertheless, it has been shown that atrial wall stretching can activate the myocardial APJ axis [164]. Moreover, APJ was found to be essential for stretch-induced contractility and may also induce ectopic electrical activity by Ca2+ sensitization of myofilaments. It is believed that apelin counteracts APJ’s stretch-triggered hypertrophy signaling by suppressing Ca2+ transients [164]. Along with it, there are a variety of vascular effects of apelin that include regulation of systolic and diastolic blood pressure through vasorelaxation and an increase in regional blood flow [165,166].
Previous studies have shown that circulating levels of apelin were sufficiently lower in patients with established cardiovascular diseases (coronary artery disease, myocardial infarction, acute coronary syndrome, HF), T2DM, and obesity than in healthy volunteers [167,168]. A meta-analysis of 30 studies revealed a negative association of apelin serum levels with cardiovascular diseases [169]. However, peripheral blood apelin concentrations were not only significantly decreased in AF patients compared with healthy controls but also independently predicted recurrent AF in patients with persistent AF. This included cases occurring after pulmonary vein isolation in subjects without structural heart disease [170,171,172]. It has been suggested that low apelin levels may interfere with AF susceptibility through elevated atrial NADPH-dependent oxidative stress and the TGF-β/Smad2/α-SMA pathway associated with mitochondrial dysfunction and myocardial fibrosis [173,174]. In addition, the apelin/APJ axis might be involved in atrial thrombus formation among AF patients, possibly as a result of concomitant downstream plasminogen activator inhibitor-1 (PAI-1) [175].
The predictive role of apelin for AF occurrence after electric cardioversion remains uncertain. In a small comparative study, Kallergis EM et al. (2010) [176] showed that baseline apelin levels did not independently predict AF recurrence, whereas NT-proBNP did. Interestingly, maintenance of sinus rhythm after electrical cardioversion resulted in an increase in serum apelin levels and a decrease in serum NT-pro-BNP levels. However, more studies are needed to clarify apelin’s discriminative potency for AF recurrence in AF patients after electrical cardioversion, with comparisons of apelin’s predictive value to other conventional and promising biomarkers.

4.4.2. Irisin

Irisin was previously described as a hormone-like myokine, which is mainly secreted by skeletal muscle and myocardium and is a derivative of the membrane protein fibronectin type III domain-containing 5 (FNDC5) [177]. Exercise increases serum levels of irisin, which exert cytoprotective effects on remote organs and tissues, including the heart, kidney, vasculature, bones, and brain [177,178]. Irisin interacts with αV/β5 integrin on the surface of target cells and induces a wide range of biological effects, including stimulation of glucose and lipid metabolism, increase in insulin resistance, browning of visceral adipose tissue, thermogenesis, angiogenesis, survival of osteoblasts, and production of bone-related proteins such as sclerostin [179,180,181,182].
Serum irisin levels were significantly decreased in obese and T2DM patients compared with nondiabetic controls, as well as in patients with known cardiovascular disease (cardiac hypertrophy, stable coronary artery disease, chronic HF, multifocal atherosclerosis) compared with healthy volunteers [183,184]. On the contrary, acute HF, acute coronary syndrome, and acute myocardial infarction were associated with an increase in irisin levels, which is considered an adaptive factor that reduces endothelial damage by inhibiting inflammatory reactions and suppressing oxidative stress [185,186,187]. A low irisin level was described as an independent predictor of clinical outcomes in HF patients [188,189]. Although patients with HFpEF and AF had significantly lower irisin levels than those without AF [190], the role of irisin in predicting AF-related events, including relapse after electric cardioversion, has not yet been investigated.

4.4.3. Bone-Related Proteins

There is growing strong evidence that inflammatory responses are involved in the development of AF and its complications. Bone-related proteins are matricellular peptides that mediate diverse biological functions and are involved in many pathological conditions in cardiovascular disease, including fibrosis, microvascular inflammation, calcification, extracellular remodeling, and atherosclerotic plaque formation [191]. Bone-related proteins, such as osteoprotegerin (OPG) and TNF-related apoptosis-inducing ligand (TRAIL), mediate a link between cardiovascular comorbidities and diseases, such as diabetes mellitus, CKD, atherosclerosis, HF, vascular calcification, and the occurrence of AF [192]. Indeed, cardiovascular comorbidities were associated with higher OPG levels and lower TRAIL levels immediately after the first hours of AF paroxysm [125,193]. Furthermore, osteopontine (OPN) levels were related to an increased risk of systemic thromboembolism and ischemic stroke in patients with AF [194]. OPG and OPN were found to be predictors of HF outcomes independent of AF presence and have been included in a multiple-scoring system to predict survival in chronic HF [195]. In a small clinical study involving 100 non-CVD patients with and without AF recurrence, low levels of bone morphogenetic protein 10 exhibited predictive value for sinus rhythm maintenance with a striking similarity to NT-proBNP [83]. However, it remains unclear whether these biomarkers have prognostic abilities for the maintenance of sinus rhythm in AF patients after electrical cardioversion.

4.5. Biomarkers of Oxidative Stress and Endothelial Dysfunction

4.5.1. Cell-Free Circulating DNA

Cell-free circulating DNA (cfcDNA) circulates in two main pools: circular and single-stranded molecules belonging to mitochondrial-derived and nuclear-derived subpopulations, reflecting patterns of DNA methylation and a variety of neutrophil extracellular traps (NETosis) [196,197]. The cfcDNA are determined in subdetectable concentrations under certain physiological conditions, such as physical exercise, whereas increased circulating levels of these fragments are strongly associated with cardiovascular, autoimmune, rheumatic diseases, infections, and malignancy [198,199,200,201,202]. The main causes of cfcDNA production are mitochondrial dysfunction and inflammation, which are powerful drivers of numerous diseases and conditions, including AF [203].
Wiersma M. et al. (2020) [204] reported that levels of cell-free circulating mitochondrial DNA (cfc-mtDNA) were significantly increased in patients with paroxysmal AF undergoing AF treatment, especially in men and in patients with AF recurrence after electrical cardioversion or pulmonary vein isolation. In contrast, cfc-mtDNA levels gradually decreased in patients with persistent AF and long-standing persistent AF. Nevertheless, the authors suggested that cfc-mtDNA levels might be associated with the stage of AF and the risk of AF recurrence after treatment, especially in men. Gender differences in descriptive values of cfc-mtDNA for AF recurrence remain poorly understood but could be related to different comorbidities in both subpopulations. However, another study found no significant changes in mtDNA copy number in the peripheral blood of AF patients of different sex and age [205]. Perhaps, cfcDNA may be included in the multiple biomarker models with the aim of improving their predictive potency in AF patients with low levels of NT-proBNP or in AF patients with malignancy who are treated with chemotherapy.

4.5.2. mRNA

MicroRNAs (miRNAs) participate in atrial remodeling and cardiac fibrosis, contributing to the development of AF [206]. Garcia-Elias A et al. (2021) [207] established that circulating levels of miR-199a-5p and miR-22-5p, which regulate fibrogenic response in the myocardium, were higher in HFrEF patients with AF than in those without AF [207]. MiR-21, which corresponds to atrial fibrosis, is associated with the risk of persistent AF in patients with left atrial enlargement [208]. Interestingly, increased circulating levels of miR-1-3p, which is a myosine gene regulator involved in hypertrophy, myocardial infarction, and cardiac arrhythmogenesis, predicted a high risk of subclinical AF [209]. MiR423, which downregulates fibrosis-related genes such as collagen I, collagen III, fibronectin, and TGF-beta, may be a pivotal factor in stratifying patients at risk of AF occurrence and persistence [210]. Moreover, differences in miRNA expression in the atrial myocardium of men and women may mediate a sex-specific association between circulating miRNAs in plasma and AF at the population level [206]. In addition, there is evidence that epigenetic regulation of NETosis may participate in the development of AF susceptibility. As a matter of fact, miR-146a and miR21 may provide prognostic information in patients with AF [211,212] due to its direct effects on NETosis. In a study by da Silva AMG (2018) [213], miR-21, miR-133b, and miR-499, which are directly involved in the downregulation of apoptosis and fibrosis, were found to be directly involved in AF. However, it remains to be determined whether a signature of mi-Rs can be used to predict poor response to AF treatment, including electrical cardioversion. At the same time, Zhou Q et al. (2018) [213] reported that among 123 miRs affecting cardiac fibrosis, hypertrophy, and inflammation by relation with the SMAD7 and FASLG genes, only miR-21 demonstrated a positive correlation with left atrial low-voltage areas in patients with persistent AF and was associated with post-ablation outcome. Overall, the signature of miRs appears to be a more promising tool for higher AF risk than for outcomes after treatment, although this conjecture needs to be further investigated in the future.

4.5.3. Asymmetric Dimethylarginine

Asymmetric dimethylarginine (ADMA) is a well-known biomarker of endothelial dysfunction that indirectly reflects vascular NO production and exhibits certain predictive information for mortality and morbidity of cardiovascular diseases, including AF [214,215]. In the population-based Gutenberg Health Study (n = 5000), ADMA levels were correlated with left ventricular hypertrophy and AF prevalence [216]. An ARISTOTLE (Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation) substudy showed that elevated ADMA levels exhibited a weak association with thromboembolic events in AF patients treated with anticoagulants (warfarin or apixaban) for a median of 1.9 years [217]. The investigators found that tertile groups of ADMA levels were sufficiently associated with death, stroke, and systemic embolism and that incorporating ADMA into CHA2DS2-VASc or HAS-BLED predictive models significantly improved C-indices for those clinical outcomes [217].
There is strong evidence that acute and persistent episodes of AF seem to show elevated ADMA levels accompanied by increased biomarkers of ischemic myocardial injury like cardiac troponins [218]. In the animal AF model, ADMA concentrations in peripheral blood returned to normal within 24 h after successful electrical cardioversion [218]. Along with it, increased circulating levels of ADMA in AF may be reduced by a Mediterranean diet and statin treatment [219,220]. Thus, being closely associated with thrombus formation and CHADS2/CHA2DS2-VASc score, ADMA is a biomarker for predicting pro-thrombotic risk in AF [221,222].
There are controversial data for ADMA’s predictive ability regarding AF recurrence after electrical cardioversion. Xia W et al. (208) [223] reported that elevated ADMA levels were strongly associated with an increased risk of AF relapse within 1 month after electrical cardioversion. On the contrary, Tveit A et al. (2010) [224] found that the levels of ADMA and the L-arginine/ADMA ratio did not exert predictive ability for sinus rhythm maintenance after electrical cardioversion, while the L-arginine/ADMA ratio remained elevated in patients with sinus rhythm for 6 months compared with patients with AF recurrence. The discriminative potency of ADMA may be strongly related to comorbidities. Indeed, serum ADMA levels were not associated with incident AF in the general population after adjusting for other cardiovascular risk factors [224]. Overall, the utility of ADMA refines clinical risk stratification in AF regardless of the treatment strategy.

5. Conclusions

Previous clinical studies demonstrated limited ability to predict the efficacy of electrical cardioversion with conventional biomarkers, which described adverse cardiac remodeling, biomechanical stress, fibrosis, inflammation, endothelial dysfunction, oxidative stress, and mitochondrial dysfunction. Epigenetic biomarkers such as miRs and biomarkers of oxidative stress and inflammation such as cfcDNA appear to show highly variable results in predicting post-procedural events. A biomarker-based strategy for predicting events after AF treatment requires extensive future investigation, especially in different gender and variable comorbidity profiles. Therefore, a multiple biomarker approach may be more useful than using a single biomarker for patients with different forms of AF. Large clinical trials are needed to make direct face-to-face comparisons with different biomarkers and their combinations.

Author Contributions

Conceptualization, O.D. and M.L.; methodology, A.E.B.; software, O.D.; formal analysis, O.D.; investigation, O.D.; resources, O.D.; data curation, A.E.B.; writing—original draft preparation, O.D., M.L. and A.E.B.; writing—review and editing, O.D., U.C.H., M.M., E.B., S.X.G., M.L. and A.E.B.; supervision, A.E.B.; project administration, O.D.; advice for analysis and interpretation of the data: U.C.H., M.M., E.B. and S.X.G. All authors have participated in drafting the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Predictive values of different biomarkers in prediction of AF-related complications after electrical cardioversion.
Table A1. Predictive values of different biomarkers in prediction of AF-related complications after electrical cardioversion.
BiomarkersPopulationObservation PeriodSignificance/OutcomesReferences
Biomechanical stress
BNP58 patients with persistent AF and preserved LVEF6 monthsBaseline BNP level and the magnitude of its decrease after successful cardioversion predicted AF recurrence[82]
NT-proBNP and BMP10100 non-CVD patients with and without AF recurrence 30-day follow-upLow NT-proBNP levels and BMP10 levels after electric cardioversion predicted sinus rhythm restoration [83]
BNP and NT-proBNP43 patients with persistent AF18 monthsPre- and post-procedural levels of BNP and NT-proBNP did not predict new episodes of AF[84]
NT-proBNP199 patients with persistent AF30 daysThe levels of NT-proBNP > 500 ng/L predicted recurrence of AF in 30 days after successful electrical cardioversion[85]
NT-pro-BNP40 patients with persistent AF1 monthElevated baseline NT-pro-BNP predicted AF recurrence after electric cardioversion[86]
NT-pro-BNP171 patients with persistent AF without HF1 monthPre-cardioversion and post-cardioversion NT-pro-BNP levels did not predict a relapse of AF in patients without HF[87]
ANP and BNP71 HF patients with persistent AF1 monthLow ANP and high BNP levels before electric cardioversion independently predicted recurrent AF[88]
ANP and BNP60 patients with persistent AF12 monthsThe BNP level ≥700 fmol/mL on day 7 after cardioversion predicted AF recurrence. ANP level was not predictive of AF recurrence[89]
NT-proBNP200 patients with newly onset AF with and without HF1 monthNT-proBNP levels of either ≤450 pg/mL or >1800 pg/mL had positive and negative predictive values for cardioversion in rate-control and rhythm-control strategies[90]
Cardiac fibrosis
Gal-390 patients with persistent AF 3 monthsSerum Gal-3 level independently predicted early AF recurrence following successful direct-current electrical cardioversion.[98]
Gal-3 82 patients with persistent AF1 monthBaseline serum levels of Gal-3 were not associated with a risk of recurrent AF[101]
Gal-375 non-HF patients with paroxysmal or persistent AF1 yearPre-procedural Gal-3 levels did not predict recurrent AF[102]
sST280 patients with persistent AF without HF12 monthsSerum levels of sST2 predict sinus rhythm maintenance after cardioversion of AF in patients without HF[112]
FGF-2379 patients with persistent AF12 monthsFGF-23, but not Gal-3, PIIINP, and ICTP, had weak predictive ability for relapsing AF[113]
PIIINP88 patients with maintenance of sinus rhythm and 54 patients with AF recurrence24 monthsBaseline PIIINP levels >0.72 U/mL independently predicted AF recurrence after electric cardioversion[114]
Inflammation
GDF15 82 patients with persistent AF1 monthGDF-15 levels correlated positively with the CHA2DS2-VASc score, but not associated with a risk of recurrent AF after electric cardioversion[101]
hs-CRP102 patients with non-valvular persistent AF1 yearLow levels of hs-CRP were associated with long-term maintenance of sinus rhythm after electrical cardioversion for AF[137]
hs-CRP53 patients with persistent AF and a mean LVEF of 58.7 ± 6%3 weeksNo changes in hs-CRP levels and decrease in NT-proBNP levels after effective cardioversion. Pre-procedural levels of hs-CRP predicted recurrence rate of AF[138]
hs-CRP57 patients with a mean LVEF of 58.7 ± 6%3 weeksPre-procedural levels of hs-CRP, but not NT-proBNP, predicted recurrence rate of AF[139]
hs-CRP60 patients who received amiodarone for sinus rhythm maintenance3 yearsPre-procedural levels of CRP >0.43 mg/dL were an independent predictor of AF recurrence[140]
hs-CRP106 patients with a history of symptomatic AF lasting ≥ 1 day36 daysPre-procedural hs-CRP levels ≥0.06 mg/dL predicted both AF recurrence and maintenance of sinus rhythm[141]
hs-CRP56 patients with persistent AF180 daysPre-procedural hs-CRP <0.8 mg/L was significantly associated with lower AF recurrence rates and maintenance of sinus rhythm[142]
hs-CRP216 patients with persistent AF12 monthsThe baseline and 2-day levels of hs-CRP levels contributed a risk of AF recurrence[145]
Apelin and NT-proBNP40 patients with persistent AF and 15 controls in sinus rhythm1 monthPre-procedural apelin levels were lower and NT-pro-BNP levels were higher in patients with AF compared to controls. Cardioversion led to an increase in apelin levels and a decrease in NT-proBNP levels. Apelin did not predict AF recurrence, but NT-proBNP did[176]
Biomarkers of oxidative stress and mitochondrial dysfunction
cfc-mtDNA59 non-AF patients undergoing cardiac surgery, 100 patients with paroxysmal AF, 116 patients with persistent AF, 20 longstanding-persistent AF individuals and 84 control individuals-Elevated cfc-mtDNA levels were found in patients with paroxysmal AF undergoing electrical cardioversion or pulmonary vein isolation, as well as in patients with AF relapse after AF treatment. In patients with persistent AF and longstanding persistent AF, the levels of cfc-mtDNA gradually decreased[204]
miR-199a-5p and miR-22-5p49 HFrEF with AF and 49 HFrEF with sinus rhythm-Elevated levels of circulating miR-199a-5p and miR-22-5p were associated with AF in HFrEF patients[207]
miR-2160 persistent AF patients and 60 matched sinus rhythm volunteers-Circulating miR-21 positively correlates with the quantification of left atrial fibrosis and is associated with the risk of persistent AF in patients with left atrial enlargement[208]
miR-1-3p64 consecutive patients with cryptogenic stroke, 9 patients with AF and 9 individuals with sinus rhythm6 and 12 monthsElevated plasma levels of miR-1-3p predicted AF [209]
miR-21, miR-133a, miR-133b, miR-150, miR-328, and miR-4995 acute new-onset AF patients, 16 well-controlled AF and 15 control-miR-21, miR-133b, and miR-499, which downregulate apoptosis and fibrosis, were found to be directly related to AF[213]
Biomarkers of endothelial dysfunction
ADMA64 patients with persistent AF1 monthHigh levels of ADMA were strongly associated with an increased risk of AF relapse after electrical cardioversion[222]
ADMA98 patients with persistent AF6 monthsChanges in ADMA did not predict rhythm outcome after electrical cardioversion[223]
Abbreviations: ADMA, asymmetric dimethylarginine; ANP, atrial natriuretic peptide; CVD, cardiovascular disease; BNP, brain natriuretic peptide; BMP10, bone morphogenetic protein 10; HF, heart failure; hs-CRP, high-sensitivity C-reactive protein; LVEF, left ventricular ejection fraction; NT-proBNP, N-terminal pro-B-type natriuretic peptide; sST2, soluble suppressor tumorigenisity-2; Gal-3, galectin-3; PIIINP, procollagen type III N terminal peptide; ICTP, type I collagen carboxyl telopeptide; FGF-23, fibroblast growth factor 23, cfc-mtDNA, cell-free circulating mitochondrial DNA.

References

  1. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the Diagnosis and Management of Atrial Fibrillation Developed in Collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the Diagnosis and Management of Atrial Fibrillation of the European Society of Cardiology (ESC) Developed with the Special Contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 2021, 42, 373–498. [Google Scholar] [CrossRef] [PubMed]
  2. Zoni-Berisso, M.; Lercari, F.; Carazza, T.; Domenicucci, S. Epidemiology of Atrial Fibrillation: European Perspective. Clin. Epidemiol. 2014, 6, 213–220. [Google Scholar] [CrossRef]
  3. Magnussen, C.; Niiranen, T.J.; Ojeda, F.M.; Gianfagna, F.; Blankenberg, S.; Njølstad, I.; Vartiainen, E.; Sans, S.; Pasterkamp, G.; Hughes, M.; et al. Sex Differences and Similarities in Atrial Fibrillation Epidemiology, Risk Factors, and Mortality in Community Cohorts: Results from the BiomarCaRE Consortium (Biomarker for Cardiovascular Risk Assessment in Europe). Circulation 2017, 136, 1588–1597. [Google Scholar] [CrossRef] [PubMed]
  4. Wachter, R. Vorhofflimmern als Komorbidität bei Herzinsuffizienz. Internist 2018, 59, 415–419. [Google Scholar] [CrossRef]
  5. Alonso, A.; Almuwaqqat, Z.; Chamberlain, A. Mortality in Atrial Fibrillation: Is It Changing? Trends Cardiovasc. Med. 2021, 31, 469–473. [Google Scholar] [CrossRef] [PubMed]
  6. Walczak-Galezewska, M.; Markowska, M.; Braszak, A.; Bryl, W.; Bogdanski, P. Atrial Fibrillation and Obesity: Should Doctors Focus on This Comorbidity? Minerva Med. 2019, 110, 175–176. [Google Scholar] [CrossRef] [PubMed]
  7. Hong, K.L.; Glover, B.M. The Impact of Lifestyle Intervention on Atrial Fibrillation. Curr. Opin. Cardiol. 2018, 33, 14–19. [Google Scholar] [CrossRef] [PubMed]
  8. Shaikh, F.; Pasch, L.B.; Newton, P.J.; Bajorek, B.V.; Ferguson, C. Addressing Multimorbidity and Polypharmacy in Individuals with Atrial Fibrillation. Curr. Cardiol. Rep. 2018, 20, 32. [Google Scholar] [CrossRef] [PubMed]
  9. Heijman, J.; Linz, D.; Schotten, U. Dynamics of Atrial Fibrillation Mechanisms and Comorbidities. Annu. Rev. Physiol. 2021, 83, 83–106. [Google Scholar] [CrossRef] [PubMed]
  10. Schoonderwoerd, B.A.; Van Gelder, I.C.; Van Veldhuisen, D.J.; Van den Berg, M.P.; Crijns, H.J.G.M. Electrical and Structural Remodeling: Role in the Genesis and Maintenance of Atrial Fibrillation. Prog. Cardiovasc. Dis. 2005, 48, 153–168. [Google Scholar] [CrossRef] [PubMed]
  11. Cha, T.-J.; Ehrlich, J.R.; Zhang, L.; Shi, Y.-F.; Tardif, J.-C.; Leung, T.K.; Nattel, S. Dissociation between Ionic Remodeling and Ability to Sustain Atrial Fibrillation during Recovery from Experimental Congestive Heart Failure. Circulation 2004, 109, 412–418. [Google Scholar] [CrossRef] [PubMed]
  12. Vanbeselaere, V.; Truyers, C.; Elli, S.; Buntinx, F.; De Witte, H.; Degryse, J.; Henrard, S.; Vaes, B. Association between Atrial Fibrillation, Anticoagulation, Risk of Cerebrovascular Events and Multimorbidity in General Practice: A Registry-Based Study. BMC Cardiovasc. Disord. 2016, 16, 61. [Google Scholar] [CrossRef]
  13. Bernard, M.L. Atrial Fibrillation and Multimorbidity. Mayo Clin. Proc. 2019, 94, 2381–2382. [Google Scholar] [CrossRef] [PubMed]
  14. Kwok, C.S.; Lip, G.Y.H. The Patient Pathway Review for Atrial Fibrillation. Crit. Pathw. Cardiol. 2022, 21, 96–102. [Google Scholar] [CrossRef] [PubMed]
  15. Furniss, S.S.; Sneyd, J.R. Safe Sedation in Modern Cardiological Practice. Heart 2015, 101, 1526–1530. [Google Scholar] [CrossRef] [PubMed]
  16. Fried, A.M.; Strout, T.D.; Perron, A.D. Electrical Cardioversion for Atrial Fibrillation in the Emergency Department: A Large Single-Center Experience. Am. J. Emerg. Med. 2021, 42, 115–120. [Google Scholar] [CrossRef]
  17. Brandes, A.; Crijns, H.J.G.M.; Rienstra, M.; Kirchhof, P.; Grove, E.L.; Pedersen, K.B.; Van Gelder, I.C. Cardioversion of Atrial Fibrillation and Atrial Flutter Revisited: Current Evidence and Practical Guidance for a Common Procedure. EP Eur. 2020, 22, 1149–1161. [Google Scholar] [CrossRef]
  18. Voskoboinik, A.; Kalman, E.; Plunkett, G.; Knott, J.; Moskovitch, J.; Sanders, P.; Kistler, P.M.; Kalman, J.M. A Comparison of Early versus Delayed Elective Electrical Cardioversion for Recurrent Episodes of Persistent Atrial Fibrillation: A Multi-Center Study. Int. J. Cardiol. 2019, 284, 33–37. [Google Scholar] [CrossRef]
  19. Yoon, M.; Yang, P.-S.; Jang, E.; Yu, H.T.; Kim, T.-H.; Uhm, J.-S.; Kim, J.-Y.; Sung, J.-H.; Pak, H.-N.; Lee, M.-H.; et al. Improved Population-Based Clinical Outcomes of Patients with Atrial Fibrillation by Compliance with the Simple ABC (Atrial Fibrillation Better Care) Pathway for Integrated Care Management: A Nationwide Cohort Study. Thromb. Haemost. 2019, 119, 1695–1703. [Google Scholar] [CrossRef]
  20. Cheung, C.C.; Nattel, S.; Macle, L.; Andrade, J.G. Management of Atrial Fibrillation in 2021: An Updated Comparison of the Current CCS/CHRS, ESC, and AHA/ACC/HRS Guidelines. Can. J. Cardiol. 2021, 37, 1607–1618. [Google Scholar] [CrossRef] [PubMed]
  21. Toufan, M.; Kazemi, B.; Molazadeh, N. The Significance of the Left Atrial Volume Index in Prediction of Atrial Fibrillation Recurrence after Electrical Cardioversion. J. Cardiovasc. Thorac. Res. 2017, 9, 54–59. [Google Scholar] [CrossRef] [PubMed]
  22. Ecker, V.; Knoery, C.; Rushworth, G.; Rudd, I.; Ortner, A.; Begley, D.; Leslie, S.J. A Review of Factors Associated with Maintenance of Sinus Rhythm after Elective Electrical Cardioversion for Atrial Fibrillation. Clin. Cardiol. 2018, 41, 862–870. [Google Scholar] [CrossRef]
  23. Conte, M.; Petraglia, L.; Cabaro, S.; Valerio, V.; Poggio, P.; Pilato, E.; Attena, E.; Russo, V.; Ferro, A.; Formisano, P.; et al. Epicardial Adipose Tissue and Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front. Cardiovasc. Med. 2022, 9, 932262. [Google Scholar] [CrossRef]
  24. Abe, I.; Teshima, Y.; Kondo, H.; Kaku, H.; Kira, S.; Ikebe, Y.; Saito, S.; Fukui, A.; Shinohara, T.; Yufu, K.; et al. Association of fibrotic remodeling and cytokines/chemokines content in epicardial adipose tissue with atrial myocardial fibrosis in patients with atrial fibrillation. Heart Rhythm 2018, 15, 1717–1727. [Google Scholar] [CrossRef] [PubMed]
  25. Michniewicz, E.; Mlodawska, E.; Lopatowska, P.; Tomaszuk-Kazberuk, A.; Malyszko, J. Patients with atrial fibrillation and coronary artery disease—Double trouble. Adv. Med. Sci. 2018, 63, 30–35. [Google Scholar] [CrossRef] [PubMed]
  26. Huang, T.; Nairn, D.; Chen, J.; Mueller-Edenborn, B.; Pilia, N.; Mayer, L.; Eichenlaub, M.; Moreno-Weidmann, Z.; Allgeier, J.; Trenk, D.; et al. Structural and electrophysiological determinants of atrial cardiomyopathy identify remodeling discrepancies between paroxysmal and persistent atrial fibrillation. Front. Cardiovasc. Med. 2023, 9, 1101152. [Google Scholar] [CrossRef]
  27. Sánchez-Quintana, D.; López-Mínguez, J.R.; Pizarro, G.; Murillo, M.; Cabrera, J.A. Triggers and anatomical substrates in the genesis and perpetuation of atrial fibrillation. Curr. Cardiol. Rev. 2012, 8, 310–326. [Google Scholar] [CrossRef] [PubMed]
  28. Gomez, J.F.; Cardona, K.; Martinez, L.; Saiz, J.; Trenor, B. Electrophysiological and structural remodeling in heart failure modulate arrhythmogenesis. 2D simulation study. PLoS ONE 2014, 9, e103273. [Google Scholar] [CrossRef]
  29. Wang, Y.; Hill, J.A. Electrophysiological remodeling in heart failure. J. Mol. Cell. Cardiol. 2010, 48, 619–632. [Google Scholar] [CrossRef]
  30. Janse, M.J. Electrophysiological changes in heart failure and their relationship to arrhythmogenesis. Cardiovasc. Res. 2004, 61, 208–217. [Google Scholar] [CrossRef]
  31. Coronel, R.; Wilders, R.; Verkerk, A.O.; Wiegerinck, R.F.; Benoist, D.; Bernus, O. Electrophysiological changes in heart failure and their implications for arrhythmogenesis. Biochim. Biophys. Acta 2013, 1832, 2432–2441. [Google Scholar] [CrossRef]
  32. Aistrup, G.L.; Balke, C.W.; Wasserstrom, J.A. Arrhythmia triggers in heart failure: The smoking gun of [Ca2+]i dysregulation. Heart Rhythm 2011, 8, 1804–1808. [Google Scholar] [CrossRef]
  33. Akar, F.G.; Rosenbaum, D.S. Transmural electrophysiological heterogeneities underlying arrhythmogenesis in heart failure. Circ. Res. 2003, 93, 638–645. [Google Scholar] [CrossRef] [PubMed]
  34. Shi, C.; Wang, X.; Dong, F.; Wang, Y.; Hui, J.; Lin, Z.; Yang, J.; Xu, Y. Temporal alterations and cellular mechanisms of transmural repolarization during progression of mouse cardiac hypertrophy and failure. Acta Physiol. 2013, 208, 95–110. [Google Scholar] [CrossRef]
  35. Yang, X.; Chen, Y.; Li, Y.; Ren, X.; Xing, Y.; Shang, H. Effects of Wenxin Keli on Cardiac Hypertrophy and Arrhythmia via Regulation of the Calcium/Calmodulin Dependent Kinase II Signaling Pathway. Biomed. Res. Int. 2017, 2017, 1569235. [Google Scholar] [CrossRef]
  36. Kuba, K.; Sato, T.; Imai, Y.; Yamaguchi, T. Apelin and Elabela/Toddler; double ligands for APJ/Apelin receptor in heart development, physiology, and pathology. Peptides 2019, 111, 62–70. [Google Scholar] [CrossRef] [PubMed]
  37. Baumgartner, C.; da Costa, B.R.; Collet, T.H.; Feller, M.; Floriani, C.; Bauer, D.C.; Cappola, A.R.; Heckbert, S.R.; Ceresini, G.; Gussekloo, J.; et al. Thyroid Function within the Normal Range, Subclinical Hypothyroidism, and the Risk of Atrial Fibrillation. Circulation 2017, 136, 2100–2116. [Google Scholar] [CrossRef] [PubMed]
  38. Song, X.; Nie, L.; Long, J.; Zhao, J.; Liu, X.; Wang, L.; Liu, D.; Wang, S.; Liu, S.; Yang, J. Hydrogen sulfide alleviates hypothyroidism-induced myocardial fibrosis in rats through stimulating autophagy and inhibiting TGF-β1/Smad2 pathway. Korean J. Physiol. Pharmacol. 2023, 27, 1–8. [Google Scholar] [CrossRef]
  39. Karam, B.S.; Chavez-Moreno, A.; Koh, W.; Akar, J.G.; Akar, F.G. Oxidative stress and inflammation as central mediators of atrial fibrillation in obesity and diabetes. Cardiovasc. Diabetol. 2017, 16, 120. [Google Scholar] [CrossRef]
  40. Chan, Y.H.; Chang, G.J.; Lai, Y.J.; Chen, W.J.; Chang, S.H.; Hung, L.M.; Kuo, C.T.; Yeh, Y.H. Atrial fibrillation and its arrhythmogenesis associated with insulin resistance. Cardiovasc. Diabetol. 2019, 18, 125. [Google Scholar] [CrossRef]
  41. Glukhov, A.V.; Fedorov, V.V.; Kalish, P.W.; Ravikumar, V.K.; Lou, Q.; Janks, D.; Schuessler, R.B.; Moazami, N.; Efimov, I.R. Conduction remodeling in human end-stage nonischemic left ventricular cardiomyopathy. Circulation 2012, 125, 1835–1847. [Google Scholar] [CrossRef]
  42. Wiegerinck, R.F.; van Veen, T.A.; Belterman, C.N.; Schumacher, C.A.; Noorman, M.; de Bakker, J.M.; Coronel, R. Transmural dispersion of refractoriness and conduction velocity is associated with heterogeneously reduced connexin-43 in a rabbit model of heart failure. Heart Rhythm 2008, 5, 1178–1185. [Google Scholar] [CrossRef]
  43. Akar, F.G.; Nass, R.D.; Hahn, S.; Cingolani, E.; Shah, M.; Hesketh, G.G.; DiSilvestre, D.; Tunin, R.S.; Kass, D.A.; Tomaselli, G.F. Dynamic changes in conduction velocity and gap junction properties during development of pacing-induced heart failure. Am. J. Physiol. Heart Circ. Physiol. 2007, 293, H1223–H1230. [Google Scholar] [CrossRef]
  44. Yan, J.; Killingsworth, C.; Walcott, G.; Zhu, Y.; Litovsky, S.; Huang, J.; Ai, X.; Pogwizd, S.M. Molecular remodeling of Cx43, but not structural remodeling, promotes arrhythmias in an arrhythmogenic canine model of nonischemic heart failure. J. Mol. Cell. Cardiol. 2021, 158, 72–81. [Google Scholar] [CrossRef]
  45. Poelzing, S.; Rosenbaum, D.S. Altered connexin43 expression produces arrhythmia substrate in heart failure. Am. J. Physiol. Heart Circ. Physiol. 2004, 287, H1762–H1770. [Google Scholar] [CrossRef] [PubMed]
  46. Vitale, E.; Rosso, R.; Lo Iacono, M.; Cristallini, C.; Giachino, C.; Rastaldo, R. Apelin-13 Increases Functional Connexin-43 through Autophagy Inhibition via AKT/mTOR Pathway in the Non-Myocytic Cell Population of the Heart. Int. J. Mol. Sci. 2022, 23, 13073. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, Y.; Qiao, X.; Zhang, L.; Li, X.; Liu, Q. Apelin-13 regulates angiotensin ii-induced Cx43 downregulation and autophagy via the AMPK/mTOR signaling pathway in HL-1 cells. Physiol. Res. 2020, 69, 813–822. [Google Scholar] [CrossRef] [PubMed]
  48. Yang, M.; Song, J.J.; Yang, X.C.; Zhong, G.Z.; Zhong, J.C. MiRNA-122-5p inhibitor abolishes angiotensin II-mediated loss of autophagy and promotion of apoptosis in rat cardiofibroblasts by modulation of the apelin-AMPK-mTOR signaling. In Vitro Cell. Dev. Biol. Anim. 2022, 58, 136–148. [Google Scholar] [CrossRef]
  49. Song, J.; Zhang, Z.; Dong, Z.; Liu, X.; Liu, Y.; Li, X.; Xu, Y.; Guo, Y.; Wang, N.; Zhang, M.; et al. MicroRNA-122-5p Aggravates Angiotensin II-Mediated Myocardial Fibrosis and Dysfunction in Hypertensive Rats by Regulating the Elabela/Apelin-APJ and ACE2-GDF15-Porimin Signaling. J. Cardiovasc. Transl. Res. 2022, 15, 535–547. [Google Scholar] [CrossRef]
  50. Beyer, C.; Tokarska, L.; Stühlinger, M.; Feuchtner, G.; Hintringer, F.; Honold, S.; Fiedler, L.; Schönbauer, M.S.; Schönbauer, R.; Plank, F. Structural Cardiac Remodeling in Atrial Fibrillation. JACC Cardiovasc. Imaging 2021, 14, 2199–2208. [Google Scholar] [CrossRef] [PubMed]
  51. Jackson, L.R., 2nd; Rathakrishnan, B.; Campbell, K.; Thomas, K.L.; Piccini, J.P.; Bahnson, T.; Stiber, J.A.; Daubert, J.P. Sinus Node Dysfunction and Atrial Fibrillation: A Reversible Phenomenon? Pacing Clin. Electrophysiol. 2017, 40, 442–450. [Google Scholar] [CrossRef] [PubMed]
  52. Elliott, J.; Mainardi, L.; Rodriguez Matas, J.F. Cellular heterogeneity and repolarisation across the atria: An in silico study. Med. Biol. Eng. Comput. 2022, 60, 3153–3168. [Google Scholar] [CrossRef]
  53. Da Costa, A.; Mourot, S.; Roméyer-Bouchard, C.; Thévenin, J.; Samuel, B.; Kihel, A.; Isaaz, K. Anatomic and electrophysiological differences between chronic and paroxysmal forms of common atrial flutter and comparison with controls. Pacing Clin. Electrophysiol. 2004, 27, 1202–1211. [Google Scholar] [CrossRef]
  54. Soulat-Dufour, L.; Lang, S.; Addetia, K.; Ederhy, S.; Adavane-Scheuble, S.; Chauvet-Droit, M.; Jean, M.L.; Nhan, P.; Ben Said, R.; Kamami, I.; et al. Restoring Sinus Rhythm Reverses Cardiac Remodeling and Reduces Valvular Regurgitation in Patients with Atrial Fibrillation. J. Am. Coll. Cardiol. 2022, 79, 951–961. [Google Scholar] [CrossRef] [PubMed]
  55. Jacquemet, V.; Henriquez, C.S. Modelling cardiac fibroblasts: Interactions with myocytes and their impact on impulse propagation. Europace 2007, 9 (Suppl. S6), vi29–vi37. [Google Scholar] [CrossRef]
  56. Duffy, H.S. Fibroblasts, myofibroblasts, and fibrosis: Fact, fiction, and the future. J. Cardiovasc. Pharmacol. 2011, 57, 373–375. [Google Scholar] [CrossRef]
  57. Rohr, S. Cardiac fibroblasts in cell culture systems: Myofibroblasts all along? J. Cardiovasc. Pharmacol. 2011, 57, 389–399. [Google Scholar] [CrossRef]
  58. Dhein, S.; Salameh, A. Remodeling of Cardiac Gap Junctional Cell-Cell Coupling. Cells 2021, 10, 2422. [Google Scholar] [CrossRef]
  59. Ravens, U.; Peyronnet, R. Electrical Remodelling in Cardiac Disease. Cells 2023, 12, 230. [Google Scholar] [CrossRef] [PubMed]
  60. Wolfes, J.; Ellermann, C.; Frommeyer, G.; Eckardt, L. Evidence-based treatment of atrial fibrillation around the globe: Comparison of the latest ESC, AHA/ACC/HRS, and CCS guidelines on the management of atrial fibrillation. Rev. Cardiovasc. Med. 2022, 23, 56. [Google Scholar] [CrossRef] [PubMed]
  61. Burton, J.H.; Vinson, D.R.; Drummond, K.; Strout, T.D.; Thode, H.C.; McInturff, J.J. Electrical cardioversion of emergency department patients with atrial fibrillation. Ann. Emerg. Med. 2004, 44, 20–30. [Google Scholar] [CrossRef]
  62. Michael, J.A.; Stiell, I.G.; Agarwal, S.; Mandavia, D.P. Cardioversion of paroxysmal atrial fibrillation in the emergency department. Ann. Emerg. Med. 1999, 33, 379–387. [Google Scholar] [CrossRef] [PubMed]
  63. Dankner, R.; Shahar, A.; Novikov, I.; Agmon, U.; Ziv, A.; Hod, H. Treatment of stable atrial fibrillation in the emergency department: A population-based comparison of electrical direct-current versus pharmacological cardioversion or conservative management. Cardiology 2009, 112, 270–278. [Google Scholar] [CrossRef]
  64. Von Besser, K.; Mills, A.M. Is discharge to home after emergency department cardioversion safe for the treatment of recent-onset atrial fibrillation? Ann. Emerg. Med. 2011, 58, 517–520. [Google Scholar] [CrossRef]
  65. Xavier Scheuermeyer, F.; Grafstein, E.; Stenstrom, R.; Innes, G.; Poureslami, I.; Sighary, M. Thirty-day outcomes of emergency department patients undergoing electrical cardioversion for atrial fibrillation or flutter. Acad. Emerg. Med. 2010, 17, 408–415. [Google Scholar] [CrossRef] [PubMed]
  66. Santini, M.; De Ferrari, G.M.; Pandozi, C.; Alboni, P.; Capucci, A.; Disertori, M.; Gaita, F.; Lombardi, F.; Maggioni, A.P.; Mugelli, A.; et al. Atrial fibrillation requiring urgent medical care. Approach and outcome in the various departments of admission. Data from the atrial Fibrillation/flutter Italian REgistry (FIRE). Ital. Heart J. 2004, 5, 205–213. [Google Scholar] [PubMed]
  67. Cohn, B.G.; Keim, S.M.; Yealy, D.M. Is emergency department cardioversion of recent-onset atrial fibrillation safe and effective? J. Emerg. Med. 2013, 45, 117–127. [Google Scholar] [CrossRef]
  68. Cristoni, L.; Tampieri, A.; Mucci, F.; Iannone, P.; Venturi, A.; Cavazza, M.; Lenzi, T. Cardioversion of acute atrial fibrillation in the short observation unit: Comparison of a protocol focused on electrical cardioversion with simple antiarrhythmic treatment. Emerg. Med. J. 2011, 28, 932–937. [Google Scholar] [CrossRef]
  69. Houghton, A.R.; Sharman, A.; Pohl, J.E. Determinants of successful direct current cardioversion for atrial fibrillation and flutter: The importance of rapid referral. Br. J. Gen. Pract. 2000, 50, 710–711. [Google Scholar]
  70. Boriani, G.; Diemberger, I.; Biffi, M.; Domenichini, G.; Martignani, C.; Valzania, C.; Branzi, A. Electrical cardioversion for persistent atrial fibrillation or atrial flutter in clinical practice: Predictors of long-term outcome. Int. J. Clin. Pract. 2007, 61, 748–756. [Google Scholar] [CrossRef] [PubMed]
  71. Larsen, M.T.; Lyngborg, K.; Pedersen, F.; Corell, P. Predictive factors of maintenance of sinus rhythm after direct current (DC) cardioversion of atrial fibrillation/atrial flutter. Ugeskr. Laeger 2005, 167, 3408–3412. [Google Scholar] [PubMed]
  72. Mittal, S.; Ayati, S.; Stein, K.M.; Schwartzman, D.; Cavlovich, D.; Tchou, P.J.; Markowitz, S.M.; Slotwiner, D.J.; Scheiner, M.A.; Lerman, B.B. Transthoracic cardioversion of atrial fibrillation: Comparison of rectilinear biphasic versus damped sine wave monophasic shocks. Circulation 2000, 101, 1282–1287. [Google Scholar] [CrossRef] [PubMed]
  73. Inácio, J.F.; da Rosa Mdos, S.; Shah, J.; Rosário, J.; Vissoci, J.R.; Manica, A.L.; Rodrigues, C.G. Monophasic and biphasic shock for transthoracic conversion of atrial fibrillation: Systematic review and network meta-analysis. Resuscitation 2016, 100, 66–75. [Google Scholar] [CrossRef]
  74. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef]
  75. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, e895–e1032. [Google Scholar] [CrossRef]
  76. Cui, K.; Huang, W.; Fan, J.; Lei, H. Midregional pro-atrial natriuretic peptide is a superior biomarker to N-terminal pro-B-type natriuretic peptide in the diagnosis of heart failure patients with preserved ejection fraction. Medicine 2018, 97, e12277. [Google Scholar] [CrossRef]
  77. Okutucu, S.; Gorenek, B. Current Recommendations on Atrial Fibrillation: A Comparison of the Recent European and Canadian Guidelines. Cardiology 2022, 147, 81–89. [Google Scholar] [CrossRef] [PubMed]
  78. Schnabel, R.B.; Larson, M.G.; Yamamoto, J.F.; Sullivan, L.M.; Pencina, M.J.; Meigs, J.B.; Tofler, G.H.; Selhub, J.; Jacques, P.F.; Wolf, P.A.; et al. Relations of biomarkers of distinct pathophysiological pathways and atrial fibrillation incidence in the community. Circulation 2010, 121, 200–207. [Google Scholar] [CrossRef] [PubMed]
  79. Fan, J.; Cao, H.; Su, L.; Ling, Z.; Liu, Z.; Lan, X.; Xu, Y.; Chen, W.; Yin, Y. NT-proBNP, but not ANP and C-reactive protein, is predictive of paroxysmal atrial fibrillation in patients undergoing pulmonary vein isolation. J. Interv. Card. Electrophysiol. 2012, 33, 93–100. [Google Scholar] [CrossRef] [PubMed]
  80. Beck-da-Silva, L.; de Bold, A.; Fraser, M.; Williams, K.; Haddad, H. Brain natriuretic peptide predicts successful cardioversion in patients with atrial fibrillation and maintenance of sinus rhythm. Can. J. Cardiol. 2004, 20, 1245–1248. [Google Scholar]
  81. Xu, X.; Tang, Y. Relationship between Brain Natriuretic Peptide and Recurrence of Atrial Fibrillation after Successful Electrical Cardioversion: An Updated Meta-Analysis. Braz. J. Cardiovasc. Surg. 2017, 32, 530–535. [Google Scholar] [CrossRef] [PubMed]
  82. Ari, H.; Binici, S.; Ari, S.; Akkaya, M.; Koca, V.; Bozat, T.; Gürdoğan, M. The predictive value of plasma brain natriuretic peptide for the recurrence of atrial fibrillation six months after external cardioversion. Turk. Kardiyol. Dern. Ars. 2008, 36, 456–460. [Google Scholar]
  83. Meyre, P.B.; Aeschbacher, S.; Blum, S.; Voellmin, G.; Kastner, P.M.; Hennings, E.; Kaufmann, B.A.; Kühne, M.; Osswald, S.; Conen, D. Biomarkers associated with rhythm status after cardioversion in patients with atrial fibrillation. Sci. Rep. 2022, 12, 1680. [Google Scholar] [CrossRef] [PubMed]
  84. Wozakowska-Kapłon, B.; Bartkowiak, R.; Grabowska, U.; Janiszewska, G. B-type natriuretic peptide level after sinus rhythm restoration in patients with persistent atrial fibrillation—clinical significance. Kardiol. Pol. 2010, 68, 781–786. [Google Scholar]
  85. Andersson, J.; Rosenqvist, M.; Tornvall, P.; Boman, K. NT-proBNP predicts maintenance of sinus rhythm after electrical cardioversion. Thromb. Res. 2015, 135, 289–291. [Google Scholar] [CrossRef]
  86. Kallergis, E.M.; Manios, E.G.; Kanoupakis, E.M.; Mavrakis, H.E.; Goudis, C.A.; Maliaraki, N.E.; Saloustros, I.G.; Milathianaki, M.E.; Chlouverakis, G.I.; Vardas, P.E. Effect of sinus rhythm restoration after electrical cardioversion on apelin and brain natriuretic Peptide prohormone levels in patients with persistent atrial fibrillation. Am. J. Cardiol. 2010, 105, 90–94. [Google Scholar] [CrossRef] [PubMed]
  87. Tveit, A.; Seljeflot, I.; Grundvold, I.; Abdelnoor, M.; Arnesen, H.; Smith, P. Candesartan, NT-proBNP and recurrence of atrial fibrillation after electrical cardioversion. Int. J. Cardiol. 2009, 131, 234–239. [Google Scholar] [CrossRef]
  88. Mabuchi, N.; Tsutamoto, T.; Maeda, K.; Kinoshita, M. Plasma cardiac natriuretic peptides as biochemical markers of recurrence of atrial fibrillation in patients with mild congestive heart failure. Jpn. Circ. J. 2000, 64, 765–771. [Google Scholar] [CrossRef]
  89. Lewicka, E.; Dudzińska-Gehrmann, J.; Dąbrowska-Kugacka, A.; Zagożdżon, P.; Stepnowska, E.; Liżewska, A.; Kozłowski, D.; Raczak, G. Plasma biomarkers as predictors of recurrence of atrial fibrillation. Pol. Arch. Med. Wewn. 2015, 125, 424–433. [Google Scholar] [CrossRef]
  90. Buccelletti, F.; Gilardi, E.; Marsiliani, D.; Carroccia, A.; Silveri, N.G.; Franceschi, F. Predictive value of NT-proBNP for cardioversion in a new onset atrial fibrillation. Eur. J. Emerg. Med. 2011, 18, 157–161. [Google Scholar] [CrossRef]
  91. Koniari, I.; Artopoulou, E.; Velissaris, D.; Ainslie, M.; Mplani, V.; Karavasili, G.; Kounis, N.; Tsigkas, G. Biomarkers in the clinical management of patients with atrial fibrillation and heart failure. J. Geriatr. Cardiol. 2021, 18, 908–951. [Google Scholar] [CrossRef]
  92. Scalise, R.F.M.; De Sarro, R.; Caracciolo, A.; Lauro, R.; Squadrito, F.; Carerj, S.; Bitto, A.; Micari, A.; Bella, G.D.; Costa, F.; et al. Fibrosis after Myocardial Infarction: An Overview on Cellular Processes, Molecular Pathways, Clinical Evaluation and Prognostic Value. Med. Sci. 2021, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  93. Kawamura, M.; Ito, H.; Onuki, T.; Miyoshi, F.; Watanabe, N.; Asano, T.; Tanno, K.; Kobayashi, Y. Candesartan decreases type III procollagen-N-peptide levels and inflammatory marker levels and maintains sinus rhythm in patients with atrial fibrillation. J. Cardiovasc. Pharmacol. 2010, 55, 511–517. [Google Scholar] [CrossRef]
  94. Clementy, N.; Piver, E.; Bisson, A.; Andre, C.; Bernard, A.; Pierre, B.; Fauchier, L.; Babuty, D. Galectin-3 in Atrial Fibrillation: Mechanisms and Therapeutic Implications. Int. J. Mol. Sci. 2018, 19, 976. [Google Scholar] [CrossRef] [PubMed]
  95. Ho, J.E.; Yin, X.; Levy, D.; Vasan, R.S.; Magnani, J.W.; Ellinor, P.T.; McManus, D.D.; Lubitz, S.A.; Larson, M.G.; Benjamin, E.J. Galectin 3 and incident atrial fibrillation in the community. Am. Heart J. 2014, 167, 729–734.e1. [Google Scholar] [CrossRef]
  96. Takemoto, Y.; Ramirez, R.J.; Yokokawa, M.; Kaur, K.; Ponce-Balbuena, D.; Sinno, M.C.; Willis, B.C.; Ghanbari, H.; Ennis, S.R.; Guerrero-Serna, G.; et al. Galectin-3 Regulates Atrial Fibrillation Remodeling and Predicts Catheter Ablation Outcomes. JACC Basic Transl. Sci. 2016, 1, 143–154. [Google Scholar] [CrossRef]
  97. Yalcin, M.U.; Gurses, K.M.; Kocyigit, D.; Canpinar, H.; Canpolat, U.; Evranos, B.; Yorgun, H.; Sahiner, M.L.; Kaya, E.B.; Hazirolan, T.; et al. The Association of Serum Galectin-3 Levels with Atrial Electrical and Structural Remodeling. J. Cardiovasc. Electrophysiol. 2015, 26, 635–640. [Google Scholar] [CrossRef]
  98. Gürses, K.M.; Yalçın, M.U.; Koçyiğit, D.; Canpınar, H.; Ateş, A.H.; Canpolat, U.; Yorgun, H.; Güç, D.; Aytemir, K. Serum galectin-3 level predicts early recurrence following successful direct-current cardioversion in persistent atrial fibrillation patients. Turk. Kardiyol. Dern. Ars. 2019, 47, 564–571. [Google Scholar] [CrossRef] [PubMed]
  99. Wałek, P.; Grabowska, U.; Cieśla, E.; Sielski, J.; Roskal-Wałek, J.; Wożakowska-Kapłon, B. Analysis of the Correlation of Galectin-3 Concentration with the Measurements of Echocardiographic Parameters Assessing Left Atrial Remodeling and Function in Patients with Persistent Atrial Fibrillation. Biomolecules 2021, 11, 1108. [Google Scholar] [CrossRef]
  100. Gong, M.; Cheung, A.; Wang, Q.S.; Li, G.; Goudis, C.A.; Bazoukis, G.; Lip, G.Y.H.; Baranchuk, A.; Korantzopoulos, P.; Letsas, K.P.; et al. Galectin-3 and risk of atrial fibrillation: A systematic review and meta-analysis. J. Clin. Lab. Anal. 2020, 34, e23104. [Google Scholar] [CrossRef]
  101. Cichoń, M.; Mizia-Szubryt, M.; Olszanecka-Glinianowicz, M.; Bożentowicz-Wikarek, M.; Owczarek, A.J.; Michalik, R.; Mizia-Stec, K. Biomarkers of left atrial overload in obese and nonobese patients with atrial fibrillation qualified for electrical cardioversion. Kardiol. Pol. 2021, 79, 269–276. [Google Scholar] [CrossRef]
  102. Pauklin, P.; Zilmer, M.; Eha, J.; Tootsi, K.; Kals, M.; Kampus, P. Markers of Inflammation, Oxidative Stress, and Fibrosis in Patients with Atrial Fibrillation. Oxid. Med. Cell. Longev. 2022, 2022, 4556671. [Google Scholar] [CrossRef] [PubMed]
  103. Saez-Maleta, R.; Merino-Merino, A.; Gundin-Menendez, S.; Salgado-Aranda, R.; Al Kassam-Martinez, D.; Pascual-Tejerina, V.; Martin-Gonzalez, J.; Garcia-Fernandez, J.; Perez-Rivera, J.A. sST2 and Galectin-3 genotyping in patients with persistent atrial fibrillation. Mol. Biol. Rep. 2021, 48, 1601–1606. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, J.H.; Han, Q.F.; Mo, D.G. The progress of the soluble suppression of tumorigenicity 2 (sST2) in atrial fibrillation. J. Interv. Card. Electrophysiol. 2022, 65, 591–592. [Google Scholar] [CrossRef] [PubMed]
  105. Fan, J.; Li, Y.; Yan, Q.; Wu, W.; Xu, P.; Liu, L.; Luan, C.; Zhang, J.; Zheng, Q.; Xue, J. Higher serum sST2 is associated with increased left atrial low-voltage areas and atrial fibrillation recurrence in patients undergoing radiofrequency ablation. J. Interv. Card. Electrophysiol. 2022, 64, 733–742. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, L.; Chen, W.; Shao, Y.; Zhang, M.; Li, Z.; Wang, Z.; Lu, Y. Association of Soluble Suppression of Tumorigenicity 2 with New-Onset Atrial Fibrillation in Acute Myocardial Infarction. Cardiology 2022, 147, 381–388. [Google Scholar] [CrossRef]
  107. Tseng, C.C.S.; Huibers, M.M.H.; van Kuik, J.; de Weger, R.A.; Vink, A.; de Jonge, N. The interleukin-33/ST2 pathway is expressed in the failing human heart and associated with pro-fibrotic remodeling of the myocardium. J. Cardiovasc. Transl. Res. 2018, 11, 15–21. [Google Scholar] [CrossRef]
  108. Zhao, L.; Li, S.; Zhang, C.; Tian, J.; Lu, A.; Bai, R.; An, J.; Greiser, A.; Huang, J.; Ma, X. Cardiovascular magnetic resonance-determined left ventricular myocardium impairment is associated with C-reactive protein and ST2 in patients with paroxysmal atrial fibrillation. J. Cardiovasc. Magn. Reson. 2021, 23, 30. [Google Scholar] [CrossRef] [PubMed]
  109. Sun, W.P.; Du, X.; Chen, J.J. Biomarkers for Predicting the Occurrence and Progression of Atrial Fibrillation: Soluble Suppression of Tumorigenicity 2 Protein and Tissue Inhibitor of Matrix Metalloproteinase-1. Int. J. Clin. Pract. 2022, 2022, 6926510. [Google Scholar] [CrossRef] [PubMed]
  110. Okar, S.; Kaypakli, O.; Şahin, D.Y.; Koç, M. Fibrosis Marker Soluble ST2 Predicts Atrial Fibrillation Recurrence after Cryoballoon Catheter Ablation of Nonvalvular Paroxysmal Atrial Fibrillation. Korean Circ. J. 2018, 48, 920–929. [Google Scholar] [CrossRef]
  111. Liu, H.; Wang, K.; Lin, Y.; Liang, X.; Zhao, S.; Li, M.; Chen, M. Role of sST2 in predicting recurrence of atrial fibrillation after radiofrequency catheter ablation. Pacing Clin. Electrophysiol. 2020, 43, 1235–1241. [Google Scholar] [CrossRef]
  112. Wałek, P.; Gorczyca, I.; Grabowska, U.; Spałek, M.; Wożakowska-Kapłon, B. The prognostic value of soluble suppression of tumourigenicity 2 and galectin-3 for sinus rhythm maintenance after cardioversion due to persistent atrial fibrillation in patients with normal left ventricular systolic function. Europace 2020, 22, 1470–1479. [Google Scholar] [CrossRef]
  113. Begg, G.A.; Lip, G.Y.; Plein, S.; Tayebjee, M.H. Circulating biomarkers of fibrosis and cardioversion of atrial fibrillation: A prospective, controlled cohort study. Clin. Biochem. 2017, 50, 11–15. [Google Scholar] [CrossRef] [PubMed]
  114. Kawamura, M.; Munetsugu, Y.; Kawasaki, S.; Onishi, K.; Onuma, Y.; Kikuchi, M.; Tanno, K.; Kobayashi, Y. Type III procollagen-N-peptide as a predictor of persistent atrial fibrillation recurrence after cardioversion. Europace 2012, 14, 1719–1725. [Google Scholar] [CrossRef] [PubMed]
  115. Çöteli, C.; Hazırolan, T.; Aytemir, K.; Erdemir, A.G.; Bakır, E.N.; Canpolat, U.; Yorgun, H.; Ateş, A.H.; Kaya, E.B.; Dikmen, Z.G.; et al. Evaluation of atrial fibrosis in atrial fibrillation patients with three different methods. Turk. J. Med. Sci. 2022, 52, 175–187. [Google Scholar] [CrossRef]
  116. Dong, Q.; Li, S.; Wang, W.; Han, L.; Xia, Z.; Wu, Y.; Tang, Y.; Li, J.; Cheng, X. FGF23 regulates atrial fibrosis in atrial fibrillation by mediating the STAT3 and SMAD3 pathways. J. Cell. Physiol. 2019, 234, 19502–19510. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, J.M.; Zhong, Y.T.; Tu, C.; Lan, J. Significance of serum fibroblast growth factor-23 and miR-208b in pathogenesis of atrial fibrillation and their relationship with prognosis. World J. Clin. Cases 2020, 8, 3458–3464. [Google Scholar] [CrossRef]
  118. Mizia-Stec, K.; Wieczorek, J.; Polak, M.; Wybraniec, M.T.; Woźniak-Skowerska, I.; Hoffmann, A.; Nowak, S.; Wikarek, M.; Wnuk-Wojnar, A.; Chudek, J.; et al. Lower soluble Klotho and higher fibroblast growth factor 23 serum levels are associated with episodes of atrial fibrillation. Cytokine 2018, 111, 106–111. [Google Scholar] [CrossRef]
  119. Chua, W.; Purmah, Y.; Cardoso, V.R.; Gkoutos, G.V.; Tull, S.P.; Neculau, G.; Thomas, M.R.; Kotecha, D.; Lip, G.Y.H.; Kirchhof, P.; et al. Data-driven discovery and validation of circulating blood-based biomarkers associated with prevalent atrial fibrillation. Eur. Heart J. 2019, 40, 1268–1276. [Google Scholar] [CrossRef]
  120. Tan, Z.; Song, T.; Huang, S.; Liu, M.; Ma, J.; Zhang, J.; Yu, P.; Liu, X. Relationship between serum growth differentiation factor 15, fibroblast growth factor-23 and risk of atrial fibrillation: A systematic review and meta-analysis. Front. Cardiovasc. Med. 2022, 9, 899667. [Google Scholar] [CrossRef]
  121. Wang, D.; Day, E.A.; Townsend, L.K.; Djordjevic, D.; Jørgensen, S.B.; Steinberg, G.R. GDF15: Emerging biology and therapeutic applications for obesity and cardiometabolic disease. Nat. Rev. Endocrinol. 2021, 17, 592–607. [Google Scholar] [CrossRef]
  122. Breit, S.N.; Brown, D.A.; Tsai, V.W. The GDF15-GFRAL Pathway in Health and Metabolic Disease: Friend or Foe? Annu. Rev. Physiol. 2021, 83, 127–151. [Google Scholar] [CrossRef]
  123. Perrone, M.A.; Aimo, A.; Bernardini, S.; Clerico, A. Inflammageing and Cardiovascular System: Focus on Cardiokines and Cardiac-Specific Biomarkers. Int. J. Mol. Sci. 2023, 24, 844. [Google Scholar] [CrossRef]
  124. Aulin, J.; Hijazi, Z.; Lindbäck, J.; Alexander, J.H.; Gersh, B.J.; Granger, C.B.; Hanna, M.; Horowitz, J.; Lopes, R.D.; McMurray, J.J.V.; et al. Biomarkers and heart failure events in patients with atrial fibrillation in the ARISTOTLE trial evaluated by a multi-state model. Am. Heart J. 2022, 251, 13–24. [Google Scholar] [CrossRef] [PubMed]
  125. Hijazi, Z.; Wallentin, L.; Lindbäck, J.; Alexander, J.H.; Connolly, S.J.; Eikelboom, J.W.; Ezekowitz, M.D.; Granger, C.B.; Lopes, R.D.; Pol, T.; et al. Screening of Multiple Biomarkers Associated with Ischemic Stroke in Atrial Fibrillation. J. Am. Heart Assoc. 2020, 9, e018984. [Google Scholar] [CrossRef]
  126. Charafeddine, K.; Zakka, P.; Bou Dargham, B.; Abdulhai, F.; Zakka, K.; Zouein, F.A.; Refaat, M. Potential Biomarkers in Atrial Fibrillation: Insight into Their Clinical Significance. J. Cardiovasc. Pharmacol. 2021, 78, 184–191. [Google Scholar] [CrossRef] [PubMed]
  127. Shao, Q.; Liu, H.; Ng, C.Y.; Xu, G.; Liu, E.; Li, G.; Liu, T. Circulating serum levels of growth differentiation factor-15 and neuregulin-1 in patients with paroxysmal non-valvular atrial fibrillation. Int. J. Cardiol. 2014, 172, e311–e313. [Google Scholar] [CrossRef]
  128. Eddy, A.C.; Trask, A.J. Growth differentiation factor-15 and its role in diabetes and cardiovascular disease. Cytokine Growth Factor Rev. 2021, 57, 11–18. [Google Scholar] [CrossRef] [PubMed]
  129. Lyngbakken, M.N.; Rønningen, P.S.; Solberg, M.G.; Berge, T.; Brynildsen, J.; Aagaard, E.N.; Kvisvik, B.; Røsjø, H.; Steine, K.; Tveit, A.; et al. Prediction of incident atrial fibrillation with cardiac biomarkers and left atrial volumes. Heart 2023, 109, 356–363. [Google Scholar] [CrossRef] [PubMed]
  130. Matusik, P.T.; Małecka, B.; Lelakowski, J.; Undas, A. Association of NT-proBNP and GDF-15 with markers of a prothrombotic state in patients with atrial fibrillation off anticoagulation. Clin. Res. Cardiol. 2020, 109, 426–434. [Google Scholar] [CrossRef]
  131. Ding, B.; Liu, P.; Zhang, F.; Hui, J.; He, L. Predicting Values of Neutrophil-to-Lymphocyte Ratio (NLR), High-Sensitivity C-Reactive Protein (hs-CRP), and Left Atrial Diameter (LAD) in Patients with Nonvalvular Atrial Fibrillation Recurrence After Radiofrequency Ablation. Med. Sci. Monit. 2022, 28, e934569. [Google Scholar] [CrossRef]
  132. Li, X.; Peng, S.; Wu, X.; Guan, B.; Tse, G.; Chen, S.; Zhou, G.; Wei, Y.; Gong, C.; Lu, X.; et al. C-reactive protein and atrial fibrillation: Insights from epidemiological and Mendelian randomization studies. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1519–1527. [Google Scholar] [CrossRef]
  133. Fu, Y.; Pan, Y.; Gao, Y.; Yang, X.; Chen, M. Predictive value of CHA2DS2-VASc score combined with hs-CRP for new-onset atrial fibrillation in elderly patients with acute myocardial infarction. BMC Cardiovasc. Disord. 2021, 21, 175. [Google Scholar] [CrossRef]
  134. Sinning, C.; Kempf, T.; Schwarzl, M.; Lanfermann, S.; Ojeda, F.; Schnabel, R.B.; Zengin, E.; Wild, P.S.; Lackner, K.J.; Munzel, T.; et al. Biomarkers for characterization of heart failure–Distinction of heart failure with preserved and reduced ejection fraction. Int. J. Cardiol. 2017, 227, 272–277. [Google Scholar] [CrossRef] [PubMed]
  135. Georgakopoulos, C.; Vlachopoulos, C.; Lazaros, G.; Tousoulis, D. Biomarkers of Atrial Fibrillation in Metabolic Syndrome. Curr. Med. Chem. 2019, 26, 898–908. [Google Scholar] [CrossRef]
  136. Sun, J.; Xu, J.; Yang, Q. Expression and predictive value of NLRP3 in patients with atrial fibrillation and stroke. Am. J. Transl. Res. 2022, 14, 3104–3112. [Google Scholar] [PubMed]
  137. Loricchio, M.L.; Cianfrocca, C.; Pasceri, V.; Bianconi, L.; Auriti, A.; Calo, L.; Lamberti, F.; Castro, A.; Pandozi, C.; Palamara, A.; et al. Relation of C-reactive protein to long-term risk of recurrence of atrial fibrillation after electrical cardioversion. Am. J. Cardiol. 2007, 99, 1421–1424. [Google Scholar] [CrossRef]
  138. Lombardi, F.; Tundo, F.; Belletti, S.; Mantero, A.; Melzi D’eril, G.V. C-reactive protein but not atrial dysfunction predicts recurrences of atrial fibrillation after cardioversion in patients with preserved left ventricular function. J. Cardiovasc. Med. 2008, 9, 581–588. [Google Scholar] [CrossRef] [PubMed]
  139. Barassi, A.; Pezzilli, R.; Morselli-Labate, A.M.; Lombardi, F.; Belletti, S.; Dogliotti, G.; Corsi, M.M.; Merlini, G.; Melzi d’Eril, G.V. Serum amyloid a and C-reactive protein independently predict the recurrences of atrial fibrillation after cardioversion in patients with preserved left ventricular function. Can. J. Cardiol. 2012, 28, 537–541. [Google Scholar] [CrossRef]
  140. Korantzopoulos, P.; Kalantzi, K.; Siogas, K.; Goudevenos, J.A. Long-term prognostic value of baseline C-reactive protein in predicting recurrence of atrial fibrillation after electrical cardioversion. Pacing Clin. Electrophysiol. 2008, 31, 1272–1276. [Google Scholar] [CrossRef] [PubMed]
  141. Watanabe, E.; Arakawa, T.; Uchiyama, T.; Kodama, I.; Hishida, H. High-sensitivity C-reactive protein is predictive of successful cardioversion for atrial fibrillation and maintenance of sinus rhythm after conversion. Int. J. Cardiol. 2006, 108, 346–353. [Google Scholar] [CrossRef]
  142. Henningsen, K.M.; Therkelsen, S.K.; Bruunsgaard, H.; Krabbe, K.S.; Pedersen, B.K.; Svendsen, J.H. Prognostic impact of hs-CRP and IL-6 in patients with persistent atrial fibrillation treated with electrical cardioversion. Scand. J. Clin. Lab. Investig. 2009, 69, 425–432. [Google Scholar] [CrossRef]
  143. Liu, T.; Li, L.; Korantzopoulos, P.; Goudevenos, J.A.; Li, G. Meta-analysis of association between C-reactive protein and immediate success of electrical cardioversion in persistent atrial fibrillation. Am. J. Cardiol. 2008, 101, 1749–1752. [Google Scholar] [CrossRef] [PubMed]
  144. Yo, C.H.; Lee, S.H.; Chang, S.S.; Lee, M.C.; Lee, C.C. Value of high-sensitivity C-reactive protein assays in predicting atrial fibrillation recurrence: A systematic review and meta-analysis. BMJ Open 2014, 4, e004418. [Google Scholar] [CrossRef]
  145. Celebi, O.O.; Celebi, S.; Canbay, A.; Ergun, G.; Aydogdu, S.; Diker, E. The effect of sinus rhythm restoration on high-sensitivity C-reactive protein levels and their association with long-term atrial fibrillation recurrence after electrical cardioversion. Cardiology 2011, 118, 168–174. [Google Scholar] [CrossRef]
  146. Krishnan, A.; Chilton, E.; Raman, J.; Saxena, P.; McFarlane, C.; Trollope, A.F.; Kinobe, R.; Chilton, L. Are Interactions between Epicardial Adipose Tissue, Cardiac Fibroblasts and Cardiac Myocytes Instrumental in Atrial Fibrosis and Atrial Fibrillation? Cells 2021, 10, 2501. [Google Scholar] [CrossRef] [PubMed]
  147. Berezin, A.E.; Berezin, A.A.; Lichtenauer, M. Myokines and Heart Failure: Challenging Role in Adverse Cardiac Remodeling, Myopathy, and Clinical Outcomes. Dis. Markers 2021, 2021, 6644631. [Google Scholar] [CrossRef] [PubMed]
  148. Suffee, N.; Moore-Morris, T.; Jagla, B.; Mougenot, N.; Dilanian, G.; Berthet, M.; Proukhnitzky, J.; Le Prince, P.; Tregouet, D.A.; Pucéat, M.; et al. Reactivation of the Epicardium at the Origin of Myocardial Fibro-Fatty Infiltration During the Atrial Cardiomyopathy. Circ. Res. 2020, 126, 1330–1342. [Google Scholar] [CrossRef] [PubMed]
  149. Fujita, K.; Maeda, N.; Sonoda, M.; Ohashi, K.; Hibuse, T.; Nishizawa, H.; Nishida, M.; Hiuge, A.; Kurata, A.; Kihara, S.; et al. Adiponectin protects against angiotensin II-induced cardiac fibrosis through activation of PPAR-alpha. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 863–870. [Google Scholar] [CrossRef]
  150. Kim, Y.; Lim, J.H.; Kim, E.N.; Hong, Y.A.; Park, H.J.; Chung, S.; Choi, B.S.; Kim, Y.S.; Park, J.Y.; Kim, H.W.; et al. Adiponectin receptor agonist ameliorates cardiac lipotoxicity via enhancing ceramide metabolism in type 2 diabetic mice. Cell Death Dis. 2022, 13, 282. [Google Scholar] [CrossRef] [PubMed]
  151. Singh, R.; Kaundal, R.K.; Zhao, B.; Bouchareb, R.; Lebeche, D. Resistin induces cardiac fibroblast-myofibroblast differentiation through JAK/STAT3 and JNK/c-Jun signaling. Pharmacol. Res. 2021, 167, 105414. [Google Scholar] [CrossRef] [PubMed]
  152. Martínez-Martínez, E.; Jurado-López, R.; Valero-Muñoz, M.; Bartolomé, M.V.; Ballesteros, S.; Luaces, M.; Briones, A.M.; López-Andrés, N.; Miana, M.; Cachofeiro, V. Leptin induces cardiac fibrosis through galectin-3, mTOR and oxidative stress: Potential role in obesity. J. Hypertens. 2014, 32, 1104–1114; discussion 1114. [Google Scholar] [CrossRef]
  153. Rienstra, M.; Sun, J.X.; Lubitz, S.A.; Frankel, D.S.; Vasan, R.S.; Levy, D.; Magnani, J.W.; Sullivan, L.M.; Meigs, J.B.; Ellinor, P.T.; et al. Plasma resistin, adiponectin, and risk of incident atrial fibrillation: The Framingham Offspring Study. Am. Heart J. 2012, 163, 119–124.e1. [Google Scholar] [CrossRef]
  154. Peller, M.; Kapłon-Cieślicka, A.; Rosiak, M.; Tymińska, A.; Ozierański, K.; Eyileten, C.; Kondracka, A.; Mirowska-Guzel, D.; Opolski, G.; Postuła, M.; et al. Are adipokines associated with atrial fibrillation in type 2 diabetes? Endokrynol. Pol. 2020, 71, 34–41. [Google Scholar] [CrossRef] [PubMed]
  155. Velliou, M.; Sanidas, E.; Papadopoulos, D.; Iliopoulos, D.; Mantzourani, M.; Toutouzas, K.; Barbetseas, J. Adipokines and atrial fibrillation: The important role of apelin. Hell. J. Cardiol. 2021, 62, 89–91. [Google Scholar] [CrossRef] [PubMed]
  156. Agbaedeng, T.A.; Zacharia, A.L.; Iroga, P.E.; Rathnasekara, V.M.; Munawar, D.A.; Bursill, C.; Noubiap, J.J. Associations between adipokines and atrial fibrillation: A systematic review and meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 853–862. [Google Scholar] [CrossRef] [PubMed]
  157. Antushevich, H.; Wójcik, M. Review: Apelin in disease. Clin. Chim. Acta 2018, 483, 241–248. [Google Scholar] [CrossRef] [PubMed]
  158. Ilaghi, M.; Soltanizadeh, A.; Amiri, S.; Kohlmeier, K.A.; Shabani, M. The apelin/APJ signaling system and cytoprotection: Role of its cross-talk with kappa opioid receptor. Eur. J. Pharmacol. 2022, 936, 175353. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, J.; Liu, M.; Chen, L. Novel pathogenesis: Regulation of apoptosis by Apelin/APJ system. Acta Biochim. Biophys. Sin. 2017, 49, 471–478. [Google Scholar] [CrossRef] [PubMed]
  160. Lu, L.; Wu, D.; Li, L.; Chen, L. Apelin/APJ system: A bifunctional target for cardiac hypertrophy. Int. J. Cardiol. 2017, 230, 164–170. [Google Scholar] [CrossRef]
  161. Rikitake, Y. The apelin/APJ system in the regulation of vascular tone: Friend or foe? J. Biochem. 2021, 169, 383–386. [Google Scholar] [CrossRef] [PubMed]
  162. Li, C.; Cheng, H.; Adhikari, B.K.; Wang, S.; Yang, N.; Liu, W.; Sun, J.; Wang, Y. The Role of Apelin-APJ System in Diabetes and Obesity. Front. Endocrinol. 2022, 13, 820002. [Google Scholar] [CrossRef]
  163. Parikh, V.N.; Liu, J.; Shang, C.; Woods, C.; Chang, A.C.; Zhao, M.; Charo, D.N.; Grunwald, Z.; Huang, Y.; Seo, K.; et al. Apelin and APJ orchestrate complex tissue-specific control of cardiomyocyte hypertrophy and contractility in the hypertrophy-heart failure transition. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H348–H356. [Google Scholar] [CrossRef] [PubMed]
  164. Mughal, A.; O’Rourke, S.T. Vascular effects of apelin: Mechanisms and therapeutic potential. Pharmacol. Ther. 2018, 190, 139–147. [Google Scholar] [CrossRef] [PubMed]
  165. Charles, C.J. Putative role for apelin in pressure/volume homeostasis and cardiovascular disease. Cardiovasc. Hematol. Agents Med. Chem. 2007, 5, 1–10. [Google Scholar] [CrossRef]
  166. Askin, L.; Askin, H.S.; Tanrıverdi, O.; Ozyildiz, A.G.; Duman, H. Serum apelin levels and cardiovascular diseases. North Clin. Istanb. 2022, 9, 290–294. [Google Scholar] [CrossRef] [PubMed]
  167. Riazian, M.; Khorrami, E.; Alipoor, E.; Moradmand, S.; Yaseri, M.; Hosseinzadeh-Attar, M.J. Assessment of Apelin Serum Levels in Persistent Atrial Fibrillation and Coronary Artery Disease. Am. J. Med. Sci. 2016, 352, 354–359. [Google Scholar] [CrossRef] [PubMed]
  168. Akbari, H.; Hosseini-Bensenjan, M.; Salahi, S.; Moazzen, F.; Aria, H.; Manafi, A.; Hosseini, S.; Niknam, M.; Asadikaram, G. Apelin and its ratio to lipid factors are associated with cardiovascular diseases: A systematic review and meta-analysis. PLoS ONE 2022, 17, e0271899. [Google Scholar] [CrossRef]
  169. Bohm, A.; Urban, L.; Tothova, L.; Bezak, B.; Uher, T.; Musil, P.; Kyselovic, J.; Lipton, J.; Olejnik, P.; Hatala, R. Concentration of apelin inversely correlates with atrial fibrillation burden. Bratisl. Lek. Listy 2021, 122, 165–171. [Google Scholar] [CrossRef]
  170. Wang, Y.Z.; Fan, J.; Zhong, B.; Xu, Q. Apelin: A novel prognostic predictor for atrial fibrillation recurrence after pulmonary vein isolation. Medicine 2018, 97, e12580. [Google Scholar] [CrossRef]
  171. Falcone, C.; Buzzi, M.P.; D’Angelo, A.; Schirinzi, S.; Falcone, R.; Rordorf, R.; Capettini, A.C.; Landolina, M.; Storti, C.; Pelissero, G. Apelin plasma levels predict arrhythmia recurrence in patients with persistent atrial fibrillation. Int. J. Immunopathol. Pharmacol. 2010, 23, 917–925. [Google Scholar] [CrossRef]
  172. Kim, Y.M.; Lakin, R.; Zhang, H.; Liu, J.; Sachedina, A.; Singh, M.; Wilson, E.; Perez, M.; Verma, S.; Quertermous, T.; et al. Apelin increases atrial conduction velocity, refractoriness, and prevents inducibility of atrial fibrillation. JCI Insight 2020, 5, e126525. [Google Scholar] [CrossRef] [PubMed]
  173. Lv, W.; Zhang, L.; Cheng, X.; Wang, H.; Qin, W.; Zhou, X.; Tang, B. Apelin Inhibits Angiotensin II-Induced Atrial Fibrosis and Atrial Fibrillation via TGF-β1/Smad2/α-SMA Pathway. Front. Physiol. 2020, 11, 583570. [Google Scholar] [CrossRef]
  174. Cheng, H.; Chen, Y.; Li, X.; Chen, F.; Zhao, J.; Hu, J.; Shan, A.; Qiao, S.; Wei, Z.; He, G.; et al. Involvement of Apelin/APJ Axis in Thrombogenesis in Valve Heart Disease Patients with Atrial Fibrillation. Int. Heart J. 2019, 60, 145–150. [Google Scholar] [CrossRef] [PubMed]
  175. Baba, M.; Yoshida, K.; Ieda, M. Clinical Applications of Natriuretic Peptides in Heart Failure and Atrial Fibrillation. Int. J. Mol. Sci. 2019, 20, 2824. [Google Scholar] [CrossRef] [PubMed]
  176. Colaianni, G.; Cinti, S.; Colucci, S.; Grano, M. Irisin and musculoskeletal health. Ann. New York Acad. Sci. 2017, 1402, 5–9. [Google Scholar] [CrossRef] [PubMed]
  177. Kim, H.; Wrann, C.D.; Jedrychowski, M.; Vidoni, S.; Kitase, Y.; Nagano, K.; Zhou, C.; Chou, J.; Parkman, V.A.; Novick, S.J.; et al. Irisin Mediates Effects on Bone and Fat via αV Integrin Receptors. Cell 2018, 175, 1756–1768.e17. [Google Scholar] [CrossRef]
  178. Gamas, L.; Matafome, P.; Seiça, R. Irisin and Myonectin Regulation in the Insulin Resistant Muscle: Implications to Adipose Tissue: Muscle Crosstalk. J. Diabetes Res. 2015, 2015, 359159. [Google Scholar] [CrossRef]
  179. Moreno-Navarrete, J.M.; Ortega, F.; Serrano, M.; Guerra, E.; Pardo, G.; Tinahones, F.; Ricart, W.; Fernández-Real, J.M. Irisin is expressed and produced by human muscle and adipose tissue in association with obesity and insulin resistance. J. Clin. Endocrinol. Metab. 2013, 98, E769–E778. [Google Scholar] [CrossRef] [PubMed]
  180. Yang, Z.; Chen, X.; Chen, Y.; Zhao, Q. Decreased irisin secretion contributes to muscle insulin resistance in high-fat diet mice. Int. J. Clin. Exp. Pathol. 2015, 8, 6490–6497. [Google Scholar] [PubMed]
  181. Polyzos, S.A.; Anastasilakis, A.D.; Efstathiadou, Z.A.; Makras, P.; Perakakis, N.; Kountouras, J.; Mantzoros, C.S. Irisin in metabolic diseases. Endocrine 2018, 59, 260–274. [Google Scholar] [CrossRef]
  182. Shoukry, A.; Shalaby, S.M.; El-Arabi Bdeer, S.; Mahmoud, A.A.; Mousa, M.M.; Khalifa, A. Circulating serum irisin levels in obesity and type 2 diabetes mellitus. IUBMB Life 2016, 68, 544–556. [Google Scholar] [CrossRef] [PubMed]
  183. Ho, M.Y.; Wang, C.Y. Role of Irisin in Myocardial Infarction, Heart Failure, and Cardiac Hypertrophy. Cells 2021, 10, 2103. [Google Scholar] [CrossRef]
  184. Luna-Ceron, E.; González-Gil, A.M.; Elizondo-Montemayor, L. Current Insights on the Role of Irisin in Endothelial Dysfunction. Curr. Vasc. Pharmacol. 2022, 20, 205–220. [Google Scholar] [CrossRef]
  185. Hsieh, I.C.; Ho, M.Y.; Wen, M.S.; Chen, C.C.; Hsieh, M.J.; Lin, C.P.; Yeh, J.K.; Tsai, M.L.; Yang, C.H.; Wu, V.C.; et al. Serum irisin levels are associated with adverse cardiovascular outcomes in patients with acute myocardial infarction. Int. J. Cardiol. 2018, 261, 12–17. [Google Scholar] [CrossRef]
  186. Abd El-Mottaleb, N.A.; Galal, H.M.; El Maghraby, K.M.; Gadallah, A.I. Serum irisin level in myocardial infarction patients with or without heart failure. Can. J. Physiol. Pharmacol. 2019, 97, 932–938. [Google Scholar] [CrossRef] [PubMed]
  187. Berezin, A.A.; Lichtenauer, M.; Boxhammer, E.; Fushtey, I.M.; Berezin, A.E. Serum Levels of Irisin Predict Cumulative Clinical Outcomes in Heart Failure Patients with Type 2 Diabetes Mellitus. Front. Physiol. 2022, 13, 922775. [Google Scholar] [CrossRef] [PubMed]
  188. Berezin, A.A.; Lichtenauer, M.; Boxhammer, E.; Stöhr, E.; Berezin, A.E. Discriminative Value of Serum Irisin in Prediction of Heart Failure with Different Phenotypes among Patients with Type 2 Diabetes Mellitus. Cells 2022, 11, 2794. [Google Scholar] [CrossRef] [PubMed]
  189. Bosanac, J.; Straus, L.; Novaković, M.; Košuta, D.; Božič Mijovski, M.; Tasič, J.; Jug, B. HFpEF and Atrial Fibrillation: The Enigmatic Interplay of Dysmetabolism, Biomarkers, and Vascular Endothelial Dysfunction. Dis. Markers 2022, 2022, 9539676. [Google Scholar] [CrossRef]
  190. Shirakawa, K.; Sano, M. Osteopontin in Cardiovascular Diseases. Biomolecules 2021, 11, 1047. [Google Scholar] [CrossRef]
  191. Berezin, A.E.; Kremzer, A.A. Circulating osteopontin as a marker of early coronary vascular calcification in type two diabetes mellitus patients with known asymptomatic coronary artery disease. Atherosclerosis 2013, 229, 475–481. [Google Scholar] [CrossRef]
  192. Rewiuk, K.; Grodzicki, T. Osteoprotegerin and TRAIL in Acute Onset of Atrial Fibrillation. Biomed. Res. Int. 2015, 2015, 259843. [Google Scholar] [CrossRef] [PubMed]
  193. Lin, M.; Bao, Y.; Du, Z.; Zhou, Y.; Zhang, N.; Lin, C.; Xie, Y.; Zhang, R.; Li, Q.; Quan, J.; et al. Plasma protein profiling analysis in patients with atrial fibrillation before and after three different ablation techniques. Front. Cardiovasc. Med. 2023, 9, 1077992. [Google Scholar] [CrossRef] [PubMed]
  194. Berezin, A.E.; Kremzer, A.A.; Martovitskaya, Y.V.; Samura, T.A.; Berezina, T.A.; Zulli, A.; Klimas, J.; Kruzliak, P. The utility of biomarker risk prediction score in patients with chronic heart failure. Int. J. Clin. Exp. Med. 2015, 8, 18255–18264. [Google Scholar] [CrossRef]
  195. Lo, Y.M.D.; Han, D.S.C.; Jiang, P.; Chiu, R.W.K. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science 2021, 372, eaaw3616. [Google Scholar] [CrossRef]
  196. Luo, H.; Wei, W.; Ye, Z.; Zheng, J.; Xu, R.H. Liquid Biopsy of Methylation Biomarkers in Cell-Free DNA. Trends Mol. Med. 2021, 27, 482–500. [Google Scholar] [CrossRef] [PubMed]
  197. Grosse, G.M.; Blume, N.; Abu-Fares, O.; Götz, F.; Ernst, J.; Leotescu, A.; Gabriel, M.M.; van Gemmeren, T.; Worthmann, H.; Lichtinghagen, R.; et al. Endogenous Deoxyribonuclease Activity and Cell-Free Deoxyribonucleic Acid in Acute Ischemic Stroke: A Cohort Study. Stroke 2022, 53, 1235–1244. [Google Scholar] [CrossRef]
  198. Berezina, T.A.; Kopytsya, M.P.; Petyunina, O.V.; Berezin, A.A.; Obradovic, Z.; Schmidbauer, L.; Lichtenauer, M.; Berezin, A.E. Lower Circulating Cell-Free Mitochondrial DNA Is Associated with Heart Failure in Type 2 Diabetes Mellitus Patients. Cardiogenetics 2023, 13, 15–30. [Google Scholar] [CrossRef]
  199. Li, X.; Hu, R.; Luo, T.; Peng, C.; Gong, L.; Hu, J.; Yang, S.; Li, Q. Serum cell-free DNA and progression of diabetic kidney disease: A prospective study. BMJ Open Diabetes Res. Care 2020, 8, e001078. [Google Scholar] [CrossRef]
  200. Gianni, C.; Palleschi, M.; Merloni, F.; Di Menna, G.; Sirico, M.; Sarti, S.; Virga, A.; Ulivi, P.; Cecconetto, L.; Mariotti, M.; et al. Cell-Free DNA Fragmentomics: A Promising Biomarker for Diagnosis, Prognosis and Prediction of Response in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 14197. [Google Scholar] [CrossRef] [PubMed]
  201. Hashimoto, T.; Ueki, S.; Kamide, Y.; Miyabe, Y.; Fukuchi, M.; Yokoyama, Y.; Furukawa, T.; Azuma, N.; Oka, N.; Takeuchi, H.; et al. Increased Circulating Cell-Free DNA in Eosinophilic Granulomatosis with Polyangiitis: Implications for Eosinophil Extracellular Traps and Immunothrombosis. Front. Immunol. 2022, 12, 801897. [Google Scholar] [CrossRef]
  202. Yamazoe, M.; Sasano, T.; Ihara, K.; Takahashi, K.; Nakamura, W.; Takahashi, N.; Komuro, H.; Hamada, S.; Furukawa, T. Sparsely methylated mitochondrial cell free DNA released from cardiomyocytes contributes to systemic inflammatory response accompanied by atrial fibrillation. Sci. Rep. 2021, 11, 5837. [Google Scholar] [CrossRef]
  203. Wiersma, M.; van Marion, D.M.S.; Bouman, E.J.; Li, J.; Zhang, D.; Ramos, K.S.; Lanters, E.A.H.; de Groot, N.M.S.; Brundel, B.J.J.M. Cell-Free Circulating Mitochondrial DNA: A Potential Blood-Based Marker for Atrial Fibrillation. Cells 2020, 9, 1159. [Google Scholar] [CrossRef]
  204. Soltész, B.; Urbancsek, R.; Pös, O.; Hajas, O.; Forgács, I.N.; Szilágyi, E.; Nagy-Baló, E.; Szemes, T.; Csanádi, Z.; Nagy, B. Quantification of peripheral whole blood, cell-free plasma and exosome encapsulated mitochondrial DNA copy numbers in patients with atrial fibrillation. J. Biotechnol. 2019, 299, 66–71. [Google Scholar] [CrossRef] [PubMed]
  205. Geurts, S.; Mens, M.M.J.; Bos, M.M.; Ikram, M.A.; Ghanbari, M.; Kavousi, M. Circulatory MicroRNAs in Plasma and Atrial Fibrillation in the General Population: The Rotterdam Study. Genes 2021, 13, 11. [Google Scholar] [CrossRef] [PubMed]
  206. Garcia-Elias, A.; Tajes, M.; Yañez-Bisbe, L.; Enjuanes, C.; Comín-Colet, J.; Serra, S.A.; Fernández-Fernández, J.M.; Aguilar-Agon, K.W.; Reilly, S.; Martí-Almor, J.; et al. Atrial Fibrillation in Heart Failure Is Associated with High Levels of Circulating microRNA-199a-5p and 22-5p and a Defective Regulation of Intracellular Calcium and Cell-to-Cell Communication. Int. J. Mol. Sci. 2021, 22, 10377. [Google Scholar] [CrossRef]
  207. Chen, H.; Zhang, F.; Zhang, Y.L.; Yang, X.C. Relationship between circulating miRNA-21, atrial fibrosis, and atrial fibrillation in patients with atrial enlargement. Ann. Palliat. Med. 2021, 10, 12742–12749. [Google Scholar] [CrossRef] [PubMed]
  208. Benito, B.; García-Elías, A.; Ois, Á.; Tajes, M.; Vallès, E.; Ble, M.; Yáñez Bisbe, L.; Giralt-Steinhauer, E.; Rodríguez-Campello, A.; Cladellas Capdevila, M.; et al. Plasma levels of miRNA-1-3p are associated with subclinical atrial fibrillation in patients with cryptogenic stroke. Rev. Esp. Cardiol. (Engl. Ed). 2022, 75, 717–726, (In English and Spanish). [Google Scholar] [CrossRef]
  209. Park, H.; Park, H.; Park, J. Circulating microRNA-423 attenuates the phosphorylation of calcium handling proteins in atrial fibrillation. Mol. Med. Rep. 2022, 25, 186. [Google Scholar] [CrossRef]
  210. Arroyo, A.B.; de Los Reyes-García, A.M.; Rivera-Caravaca, J.M.; Valledor, P.; García-Barberá, N.; Roldán, V.; Vicente, V.; Martínez, C.; González-Conejero, R. MiR-146a Regulates Neutrophil Extracellular Trap Formation That Predicts Adverse Cardiovascular Events in Patients with Atrial Fibrillation. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 892–902. [Google Scholar] [CrossRef] [PubMed]
  211. Da Silva, A.M.G.; de Araújo, J.N.G.; de Oliveira, K.M.; Novaes, A.E.M.; Lopes, M.B.; de Sousa, J.C.V.; Filho, A.A.A.; Luchessi, A.D.; de Rezende, A.A.; Hirata, M.H.; et al. Circulating miRNAs in acute new-onset atrial fibrillation and their target mRNA network. J. Cardiovasc. Electrophysiol. 2018, 29, 1159–1166. [Google Scholar] [CrossRef]
  212. Zhou, Q.; Maleck, C.; von Ungern-Sternberg, S.N.I.; Neupane, B.; Heinzmann, D.; Marquardt, J.; Duckheim, M.; Scheckenbach, C.; Stimpfle, F.; Gawaz, M.; et al. Circulating MicroRNA-21 Correlates with Left Atrial Low-Voltage Areas and Is Associated with Procedure Outcome in Patients Undergoing Atrial Fibrillation Ablation. Circ. Arrhythm. Electrophysiol. 2018, 11, e006242. [Google Scholar] [CrossRef]
  213. Rochette, L.; Lorin, J.; Zeller, M.; Guilland, J.C.; Lorgis, L.; Cottin, Y.; Vergely, C. Nitric oxide synthase inhibition and oxidative stress in cardiovascular diseases: Possible therapeutic targets? Pharmacol. Ther. 2013, 140, 239–257. [Google Scholar] [CrossRef] [PubMed]
  214. Büttner, P.; Bahls, M.; Böger, R.H.; Hindricks, G.; Thiele, H.; Schwedhelm, E.; Kornej, J. Arginine derivatives in atrial fibrillation progression phenotypes. J. Mol. Med. 2020, 98, 999–1008. [Google Scholar] [CrossRef] [PubMed]
  215. Ramuschkat, M.; Appelbaum, S.; Atzler, D.; Zeller, T.; Bauer, C.; Ojeda, F.M.; Sinning, C.R.; Hoffmann, B.; Lackner, K.J.; Böger, R.H.; et al. ADMA, subclinical changes and atrial fibrillation in the general population. Int. J. Cardiol. 2016, 203, 640–646. [Google Scholar] [CrossRef]
  216. Horowitz, J.D.; De Caterina, R.; Heresztyn, T.; Alexander, J.H.; Andersson, U.; Lopes, R.D.; Steg, P.G.; Hylek, E.M.; Mohan, P.; Hanna, M.; et al. Asymmetric and Symmetric Dimethylarginine Predict Outcomes in Patients with Atrial Fibrillation: An ARISTOTLE Substudy. J. Am. Coll. Cardiol. 2018, 72, 721–733. [Google Scholar] [CrossRef]
  217. Goette, A.; Hammwöhner, M.; Bukowska, A.; Scalera, F.; Martens-Lobenhoffer, J.; Dobrev, D.; Ravens, U.; Weinert, S.; Medunjanin, S.; Lendeckel, U.; et al. The impact of rapid atrial pacing on ADMA and endothelial NOS. Int. J. Cardiol. 2012, 154, 141–146. [Google Scholar] [CrossRef]
  218. Goni, L.; Razquin, C.; Toledo, E.; Guasch-Ferré, M.; Clish, C.B.; Babio, N.; Wittenbecher, C.; Atzeni, A.; Li, J.; Liang, L.; et al. Arginine catabolism metabolites and atrial fibrillation or heart failure risk: 2 case-control studies within the Prevención con Dieta Mediterránea (PREDIMED) trial. Am. J. Clin. Nutr. 2022, 116, 653–662. [Google Scholar] [CrossRef] [PubMed]
  219. Li, J.; Xia, W.; Feng, W.; Qu, X. Effects of rosuvastatin on serum asymmetric dimethylarginine levels and atrial structural remodeling in atrial fibrillation dogs. Pacing Clin. Electrophysiol. 2012, 35, 456–464. [Google Scholar] [CrossRef]
  220. Lao, M.C.; Liu, L.J.; Luo, C.F.; Lu, G.H.; Zhai, Y.S.; Chen, X.L.; Gao, X.R. Effect of asymmetrical dimethylarginine for predicting pro-thrombotic risk in atrial fibrillation. Zhonghua Yi Xue Za Zhi 2016, 96, 2059–2063. (In Chinese) [Google Scholar] [CrossRef] [PubMed]
  221. Chao, T.F.; Lu, T.M.; Lin, Y.J.; Tsao, H.M.; Chang, S.L.; Lo, L.W.; Hu, Y.F.; Tuan, T.C.; Hsieh, M.H.; Chen, S.A. Plasma asymmetric dimethylarginine and adverse events in patients with atrial fibrillation referred for coronary angiogram. PLoS ONE 2013, 8, e71675. [Google Scholar] [CrossRef] [PubMed]
  222. Xia, W.; Qu, X.; Yu, Y.; Zhang, X.; Feng, W.; Song, Y. Asymmetric dimethylarginine concentration and early recurrence of atrial fibrillation after electrical cardioversion. Pacing Clin. Electrophysiol. 2008, 31, 1036–1040. [Google Scholar] [CrossRef] [PubMed]
  223. Tveit, A.; Arnesen, H.; Smith, P.; Bratseth, V.; Seljeflot, I. L-arginine, asymmetric dimethylarginine and rhythm outcome after electrical cardioversion for atrial fibrillation. Cardiology 2010, 117, 176–180. [Google Scholar] [CrossRef] [PubMed]
  224. Schnabel, R.B.; Maas, R.; Wang, N.; Yin, X.; Larson, M.G.; Levy, D.; Ellinor, P.T.; Lubitz, S.A.; McManus, D.D.; Magnani, J.W.; et al. Asymmetric dimethylarginine, related arginine derivatives, and incident atrial fibrillation. Am. Heart J. 2016, 176, 100–106. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Promoting factors and plausible pathogenetic mechanisms of AF. Abbreviations: AF, atrial fibrillation; CV, cardiovascular; CKD, chronic kidney disease; SAS, sympathoadrenal system; IR, insulin resistance; HF, heart failure; RAAS, renin-angiotensin-aldosterone system.
Figure 1. Promoting factors and plausible pathogenetic mechanisms of AF. Abbreviations: AF, atrial fibrillation; CV, cardiovascular; CKD, chronic kidney disease; SAS, sympathoadrenal system; IR, insulin resistance; HF, heart failure; RAAS, renin-angiotensin-aldosterone system.
Biomedicines 11 01452 g001
Figure 2. Molecular mechanisms of AF. Abbreviations: CV, cardiovascular; CRP, C-reactive protein; ECM, extracellular matrix; FGF21, fibroblast growth factor-21; GDF15, growth differential factor-15; Gal-3, galectine-3; TGF-beta-1 transforming growth factor beta-1, TNFα tumor necrosis factor alpha; TRPC3 transient receptor potential channel-3; TRPM7, transient receptor potential cation channel, subfamily M, member 7; Nav1.5, voltage-gated sodium channel, Kv1.5, voltage-gated potassium channel; MMPs. Matrix metalloproteinases; sST2, soluble suppression of tumorigenesis-2; RANKL, tumor necrosis factor ligand superfamily member 11; OPG, osteoprotegerin.
Figure 2. Molecular mechanisms of AF. Abbreviations: CV, cardiovascular; CRP, C-reactive protein; ECM, extracellular matrix; FGF21, fibroblast growth factor-21; GDF15, growth differential factor-15; Gal-3, galectine-3; TGF-beta-1 transforming growth factor beta-1, TNFα tumor necrosis factor alpha; TRPC3 transient receptor potential channel-3; TRPM7, transient receptor potential cation channel, subfamily M, member 7; Nav1.5, voltage-gated sodium channel, Kv1.5, voltage-gated potassium channel; MMPs. Matrix metalloproteinases; sST2, soluble suppression of tumorigenesis-2; RANKL, tumor necrosis factor ligand superfamily member 11; OPG, osteoprotegerin.
Biomedicines 11 01452 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Demirel, O.; Berezin, A.E.; Mirna, M.; Boxhammer, E.; Gharibeh, S.X.; Hoppe, U.C.; Lichtenauer, M. Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion. Biomedicines 2023, 11, 1452. https://doi.org/10.3390/biomedicines11051452

AMA Style

Demirel O, Berezin AE, Mirna M, Boxhammer E, Gharibeh SX, Hoppe UC, Lichtenauer M. Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion. Biomedicines. 2023; 11(5):1452. https://doi.org/10.3390/biomedicines11051452

Chicago/Turabian Style

Demirel, Ozan, Alexander E. Berezin, Moritz Mirna, Elke Boxhammer, Sarah X. Gharibeh, Uta C. Hoppe, and Michael Lichtenauer. 2023. "Biomarkers of Atrial Fibrillation Recurrence in Patients with Paroxysmal or Persistent Atrial Fibrillation Following External Direct Current Electrical Cardioversion" Biomedicines 11, no. 5: 1452. https://doi.org/10.3390/biomedicines11051452

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop